TW201247612A - Compositions for treating kidney disorders - Google Patents

Compositions for treating kidney disorders Download PDF

Info

Publication number
TW201247612A
TW201247612A TW101100587A TW101100587A TW201247612A TW 201247612 A TW201247612 A TW 201247612A TW 101100587 A TW101100587 A TW 101100587A TW 101100587 A TW101100587 A TW 101100587A TW 201247612 A TW201247612 A TW 201247612A
Authority
TW
Taiwan
Prior art keywords
group
alkyl
nr5r6
alkynyl
compound
Prior art date
Application number
TW101100587A
Other languages
Chinese (zh)
Other versions
TWI452032B (en
Inventor
Sheng-Yung Liu
San-Bao Hwang
Original Assignee
Golden Biotechnology Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Golden Biotechnology Corp filed Critical Golden Biotechnology Corp
Publication of TW201247612A publication Critical patent/TW201247612A/en
Application granted granted Critical
Publication of TWI452032B publication Critical patent/TWI452032B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • A61K31/015Hydrocarbons carbocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biotechnology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention provides methods for treating glomerulosclerosis such as focal segmental glomerulosclerosis (FSGS) or glomerulonephritis such as immunoglobulin A nephropathy (IgAN) by cyclohexenone compounds.

Description

201247612 六、發明說明: 【發明所屬之技術領域】 本發明係關於一種腎絲球硬化症或腎絲球腎炎之治療 方法,尤心一種以環己稀鲷化合物治療如局部性腎絲球硬 化症之腎絲球硬化症或如免疫球蛋白A型腎絲球腎炎之腎 絲球腎炎之方法 本申請案係主張於2011年1月21曰美國申請第 61/4352(Π號及於2011年10月7日美國申請第6i/5449i〇號之 優先權’且其内容均併入本發明以供參酌。 【先前技術】 許多疾病或缺陷均會透過攻擊腎絲球而影響腎臟功 能。腎絲球疾病包括多種基因及環境因素所導致之許多病 症’但不外乎主要分成腎絲球硬化症及腎絲球腎炎兩大類。 腎絲球硬化症係指腎辕中之腎絲球硬化。一般係指腎 臟的微血管、㈣球、及用以從血液過濾尿液之腎臟功能 單疋知尿蛋白(尿液中量最多的蛋白質)係為腎絲球硬化 's號之。腎臟損傷會影響腎藏過濾功能,且造成蛋 白由企液滲漏至尿液中H腎絲球硬化症僅為尿蛋白 許多的成因之-。而腎臟切片可能為一判斷病人是否罹患 腎絲球硬化錢其他㈣疾狀必要鋪。腎絲球硬 疒可择別意指局部性腎絲球硬化症(FSGS)及結節性腎 3 201247612 局部性腎絲球硬化症(FSGS)係以局部腎絲球硬化及足 細胞足突扁平化(foot process effacemen)之損傷特徵來判 斷。近20年的研究顯示,患有FSGS病患其末期腎臟疾病發 生率介於13至78%之間。雖然FSGS病源及發病原因尚未清 楚,但主要應該是起因於腎絲球上皮細胞本身損害’而導 致腎絲球中複雜的反應產生,進而造成腎絲球硬化。 結節性腎絲球硬化症或毛細血管間腎小球腎炎,亦可 稱之為糖尿病腎臟病變(糖尿病腎病)或基-威二氏症 (Kimmelstiel-Wilson syndrome),係為一種進行性腎並’其 係起因於腎絲球之毛細血管病變。此病的特徵為腎病變及 瀰漫性腎絲球硬化;並起因於長時間罹患糖尿病,且各國 的主要治療方式為洗腎。 截至目前為止,雖然類固醇及免疫製劑為主要用於治 療罹患原發性FSGS之方法,但此些療法對於腎損傷發展的 治療上效果仍然有線,且因其會產生各種副作用,故治療 方法僅僅是基於經驗法則,而較少根據病理證據(請參見 Matalon, et al., Semin Nephrol, 20: 309-317, 2000; Braun, et al., Cochrane Database Syst Rev: CD003233, 2008) ° 腎絲球腎炎則意指腎臟膜組織的發炎反應,其中模組 織係具有過濾功能,而可從血液中分離廢棄物及多於流趙。 其中,最常見的腎絲球腎炎為免疫球蛋白A型腎絲球^ 腎炎(IgAN),造成此腎炎加速發展之病因仍無法預測,且 臨床上仍無法預防及治療,故被視為可能會造成腎病的,艮 性腎衰竭之主要因素之一。因此,在Ig AN病患之腎臟中, 即便其他免疫上、臨床上及病理上的因素階會造成腎衰 4 201247612 竭,但系統性τ細胞活化及淋巴球/巨噬細胞/嗜中性白血球 浸潤等不正常增加,被視為造成IgAN轉換成慢性腎衰竭的 主要過程之一(Kamei,et al., Clin. J. Am. Soc. Nephrol. 2011, 14; Chan, et al., Clin. Exp. Nephrol. 2004, 8:297-303; Chao, et al., Kidney Int. 70:283-297 (2006); , Lai, K.N., 92:263-270 (2002))。再者,氧化壓力亦為主要造 成病患中及動物模型中IgAN增加及發展的因素;曾有報導 指出,在多數的人類及實驗性腎病中(包括IgAN),活性氧 (ROS)會直接引發疾病。 雖然酷質皮質固醇(glucocorticoid steroid)已廣泛應用 在治療IgAN病患,但其對於IgAN之維持腎臟功能且減緩尿 蛋白之效果仍不清楚,且因長時間使用所導致之潛在無法 控制的抑制免疫反應之副作用,仍造成用藥上的問題。 【發明内容】 本發明之一實施態樣係在提供一種治療一主體之腎絲 球疾病(如,腎絲球硬化症或腎絲球腎炎)之方法,包括:投 予一有效劑量之一環己烯酮化合物、其醫藥上可接受之鹽 類、其代謝物、其溶劑化物、或其前趨藥物至該主體’其 中,該環己烯酮化合物係具有下列結構:201247612 VI. Description of the Invention: [Technical Field of the Invention] The present invention relates to a method for treating glomerulosclerosis or glomerulonephritis, and particularly for treating a cyclamate compound such as localized glomerulosclerosis Method for kidney glomerulosclerosis or renal glomerulonephritis such as immunoglobulin type A glomerulonephritis This application claims to be filed on January 21, 2011, US application No. 61/4352 (nicknamed and in 2011 10 US Application No. 6i/5449i, nicknamed on the 7th of the month, and its contents are incorporated herein by reference. [Prior Art] Many diseases or defects affect kidney function by attacking the kidney skein. Diseases include many diseases caused by a variety of genes and environmental factors' but are mainly divided into two categories: glomerulosclerosis and glomerulonephritis. Kidney sclerosing refers to glomerular sclerosis in the renal pelvis. Refers to the microvessels of the kidney, (4) balls, and the function of the kidneys used to filter urine from the blood. The protein (the most abundant protein in the urine) is the spheroidal hardening 's. Kidney damage affects the kidney filter. Features And cause the protein to leak into the urine. The glomerular sclerosis is only the cause of many urinary proteins. The kidney section may be a judgment of whether the patient suffers from skein of the kidney. Renal spheroidal hard sputum can be referred to as local glomerulosclerosis (FSGS) and nodular kidney 3 201247612 Local glomerulosclerosis (FSGS) is local squamous cell sclerosing and podocyte foot flattening The damage characteristics of the foot process effacemen are judged. Nearly 20 years of research have shown that the incidence of end stage renal disease in patients with FSGS is between 13 and 78%. Although the cause and pathogenesis of FSGS are not clear, but mainly It should be caused by the damage of the glomerular epithelial cells themselves, which leads to the complex reaction of the kidney spheroids, which leads to the hardening of the kidney. The nodular glomerulosclerosis or intercapillary glomerulonephritis can also be called It is a diabetic nephropathy (diabetic nephropathy) or Kimmelstiel-Wilson syndrome, which is a progressive kidney and is caused by capillary disease of the glomerulus. The disease is characterized by nephropathy. And diffuse glomerular sclerosis; and caused by diabetes for a long time, and the main treatment in each country is dialysis. Up to now, although steroids and immune preparations are mainly used to treat primary FSGS, this The therapeutic effects of these therapies on the development of renal injury are still wired, and because of the various side effects, the treatment is based only on rule of thumb and less on pathological evidence (see Matalon, et al., Semin Nephrol, 20: 309-317, 2000; Braun, et al., Cochrane Database Syst Rev: CD003233, 2008) ° Renal glomerulonephritis refers to the inflammatory response of the renal membrane tissue, in which the modular tissue has a filtering function and can be separated from the blood. Waste and more than Zhao. Among them, the most common renal glomerulonephritis is immunoglobulin A type glomerular nephritis (IgAN), the cause of this accelerated development of nephritis is still unpredictable, and it is still clinically impossible to prevent and treat, so it is considered as likely One of the main causes of kidney disease, spastic renal failure. Therefore, in the kidneys of patients with Ig AN, even if other immune, clinical, and pathological factors cause renal failure, systemic tau cell activation and lymphocytes/macrophages/neutrophils Abnormal increase in infiltration, etc., is considered to be one of the main processes leading to the conversion of IgAN into chronic renal failure (Kamei, et al., Clin. J. Am. Soc. Nephrol. 2011, 14; Chan, et al., Clin. Exp. Nephrol. 2004, 8:297-303; Chao, et al., Kidney Int. 70:283-297 (2006); , Lai, KN, 92:263-270 (2002)). Furthermore, oxidative stress is also a major cause of the increase and development of IgAN in patients and animal models; it has been reported that reactive oxygen species (ROS) are directly induced in most human and experimental kidney diseases (including IgAN). disease. Although glucocorticoid steroids have been widely used in the treatment of IgAN patients, their effects on IgAN's ability to maintain kidney function and slow down urinary protein remain unclear and potentially uncontrollable inhibition due to prolonged use. The side effects of the immune response still cause problems in medication. SUMMARY OF THE INVENTION One embodiment of the present invention provides a method for treating a kidney spheroid disease (eg, glomerulosclerosis or glomerulonephritis) of a subject, comprising: administering an effective dose of one of cycloheximide An enone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中,每一 X及Y係各自獨立為氧、nr5、或硫; 201247612 R係為氫、或烷基; 每一R,、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 NR5R6、OR5、OC(=〇)R7、c(=0)0R5、c(=o)r5、卤 素、5或6員環内酯、CVC8烷基、c2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中’5或6員環内酯、CrCe烷基、C2-C8 烯基' C2-C8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、〇C(=〇)R7、c(=o)or5、c(=o)r5、 cvc8烧基、C2-C8稀基、c2-c8块基、c3-c8環烧基、C|-C8 卤烧基、及C丨-C8燒氧基之取代基所取代; 每一R·5、及R6係各自獨立為氫、或c丨-C8烷基; 係Ci-C8烷基、〇R5、或NR5R6 ; m為1-12 ;以及 Π 為 1 -12。 本發明之另一實施態樣係在提供一種減緩一主體之腎 功能損害或腎絲球損傷之方法,包括:投予一有效劑量之 一環己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、 其溶劑化物、或其前趨藥物至該主趙,其中,該環己稀酮 化合物係具有下列結構:Wherein each of the X and Y systems is independently oxygen, nr5, or sulfur; 201247612 R is hydrogen or an alkyl group; each of R, R2, and R3 is independently hydrogen, methyl, or (CH2) m-CH3; R4 is NR5R6, OR5, OC(=〇)R7, c(=0)0R5, c(=o)r5, halogen, 5 or 6 membered ring lactone, CVC8 alkyl, c2-C8 olefin a C2-C8 alkynyl, aryl, or glucosyl group, wherein '5 or 6 membered ring lactone, CrCe alkyl, C2-C8 alkenyl' C2-C8 alkynyl, aryl, and glucosyl selective One or more selected from the group consisting of NR5R6, OR5, 〇C(=〇)R7, c(=o)or5, c(=o)r5, cvc8 alkyl, C2-C8 dilute, c2-c8 block, c3 Substituting -c8 cycloalkyl, C|-C8 haloalkyl, and C丨-C8 alkoxy substituent; each R·5, and R6 are each independently hydrogen or c丨-C8 alkyl; Is a Ci-C8 alkyl group, hydrazine R5, or NR5R6; m is 1-12; and Π is 1 -12. Another embodiment of the present invention provides a method of slowing renal function damage or renal pelvic damage in a subject, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite, a solvate thereof, or a prodrug thereof to the main genus, wherein the cyclohexanone compound has the following structure:

其中,每一X及γ係各自獨立為氧、NR5 '或硫; R係為氫、或C(=0)C|-C8烧基; 每一R,、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; 6 201247612 Κ·4係為 NR_5R6、〇R5 ' 〇c(=0)R7、C!(=〇)〇R5、c(=o)r5、南 素、5或6員環内酯、CrCs烷基、c2-c8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、C「C8烷基、C2-C8 稀基、C2.-C8块基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6,> 〇R5、〇C(=〇)R7、C(=0)0R5、c(=o)r5、 C1-C8烧基、C2-C8稀基、C2-C8炔基、C3-C8環炫基、Ci-〇8 鹵烷基、及crc8烷氧基之取代基所取代; 每一r5、及汉6係各自獨立為氫、或c「c8烷基; 心係心-仏烷基、or5、或nr5r6 ; m為1-12;以及 η 為 1-12。 本發明之另一實施態樣係在提供一種提升一主體之腎 細胞核因數-Ε2相關因數2 (Nrf2)活性之方法,包括:投予 一有效劑量之一環己稀酮化合物、其醫藥上可接受之鹽 類、其代謝物、其溶劑化物、或其前趨藥物至該主體,其 中,該環己稀酮化合物係具有下列結構:Wherein each of the X and γ systems is independently oxygen, NR5 ' or sulfur; R is hydrogen, or C(=0)C|-C8 alkyl; each R, R2, and R3 are each independently hydrogen , methyl, or (CH2)m_CH3 ; 6 201247612 Κ·4 is NR_5R6, 〇R5 ' 〇c(=0)R7, C!(=〇)〇R5, c(=o)r5, Nansu, 5 Or 6-membered ring lactone, CrCs alkyl, c2-c8 alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactones, C "C8 alkyl, C2-C8" One or more selected from the group consisting of C2.-C8, aryl, and glucosyl are selected from nr5r6, > 〇R5, 〇C(=〇)R7, C(=0)0R5, c(= o) substituted with a substituent of r5, C1-C8 alkyl, C2-C8 dilute, C2-C8 alkynyl, C3-C8 cyclohexyl, Ci-〇8 haloalkyl, and crc8 alkoxy; Each of r5 and Han 6 is independently hydrogen or c "c8 alkyl; cardiac cardio-alkyl, or5, or nr5r6; m is 1-12; and η is 1-12. Another embodiment of the present invention The method provides a method for increasing the activity of a renal cell nuclear factor-Ε2 correlation factor 2 (Nrf2) of a subject, comprising: administering an effective dose of one of the cyclohexanone compounds, and medicinally Acceptable salts of the type, a metabolite thereof, a solvate thereof, or the precursor of the drug to the body, wherein the cyclohexene compound is one having the following structure:

其中,每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CPCOCVCs烷基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、OR5、〇C(=0)R7、C(=〇)〇R5、c(=〇)R5、鹵 素、5或6員環内醋、C〗-C8烧基、〇:2-(:8稀基、C2-C8炔基、 201247612 芳基、或葡萄糖基,其中,5或6員環内酯、Ci-Cs烷基、C2-C8 烯基、C2-C8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、 c】-c8烷基、C2-C8烯基、C2-C8炔基、(:3-(:8環烷基、c,-c8 鹵烷基、及crc8烷氧基之取代基所取代; 每一R5、及R6係各自獨立為氫、或c,-c8烷基; R7係 Cj-Cg炫基、OR5、或 NR5R6 ; m為1-12 ;以及 η為 1-12 。 本發明之另一實施態樣係在提供一種抑制一主體之腎 NF-κΒ活性及/或轉化生長因子(TGFhpi蛋白表現之方法, 包括:投予一有效劑量之一環己烯酮化合物、其醫藥上可 接受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該 主體,其中,該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, NR5, or sulfur; R is hydrogen or CPCOCVCs alkyl; each R丨, R2, and R3 are each independently hydrogen, methyl, or (CH2) m_CH3 ; R4 is NR5R6, OR5, 〇C(=0)R7, C(=〇)〇R5, c(=〇)R5, halogen, 5 or 6 member ring vinegar, C〗-C8 alkyl, 〇 : 2-(:8 dibasic, C2-C8 alkynyl, 201247612 aryl, or glucosyl, wherein 5 or 6 membered ring lactone, Ci-Cs alkyl, C2-C8 alkenyl, C2-C8 alkynyl , aryl, and glucosyl optionally one or more selected from the group consisting of NR5R6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, c]-c8 alkyl, C2 -C8 alkenyl, C2-C8 alkynyl, (3-(:8-cycloalkyl, c,-c8 haloalkyl, and crc8 alkoxy substituents are substituted; each R5, and R6 are each independently Is hydrogen, or c, -c8 alkyl; R7 is Cj-Cg danyl, OR5, or NR5R6; m is 1-12; and η is 1-12. Another embodiment of the present invention provides inhibition A method for expressing NF-κΒ activity and/or transforming growth factor (TGFhpi protein) in a subject, comprising: administering an effective dose of one of cyclohexenone compounds, A pharmaceutically acceptable salt, a metabolite thereof, a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中,每一 X及Y係各自獨立為氧、Nr5、或硫; R係為氫、或烷基; 每一R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 NR5r6、〇r5、〇c(=〇)R7、c(=〇)〇R5、c(=〇)R5、齒 =、5或6員環内醋、C〗_C8貌基、C2_c8稀基、C2_c8块基、 芳基、或葡萄糖基,其中,5或6員環内酯、C| C8烷基、C2 C8 烯基、C2-Cs炔基、芳基、及葡萄糖基係選擇性的經一種或 8 201247612 以上選自由 nr5r6、〇r5、oc(=o)r7、c(=〇)〇R5、c(=0)R5、 C|-C8烧基、C2-C8缔基、C2-C8块基、c3-C8環炫基、Ci-C8 齒炫•基、及C丨-C8坑氧基之取代基所取代; 每一R5、及R6係各自獨立為氫、或CrC8烷基; RACVCs烷基、OR5、或NR5R6 ; m為1 -12 ;以及 η為 1-12 。 本發明之另一實施態樣係在提供一種抑制一主體之 ROS/NO及/或p47ph°x之方法,包括:投予一有效劑量之一環 己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、其溶 劑化物、或其前趨藥物至該主體,其中,該環己烯酮化合 物係具有下列結構:Wherein each X and Y system is independently oxygen, Nr5, or sulfur; R is hydrogen or an alkyl group; each R丨, R2, and R3 are each independently hydrogen, methyl, or (CH2)m -CH3 ; R4 is NR5r6, 〇r5, 〇c(=〇)R7, c(=〇)〇R5, c(=〇)R5, tooth=, 5 or 6 member ring vinegar, C〗 _C8 appearance , C2_c8 dilute group, C2_c8 block group, aryl group, or glucosyl group, wherein 5 or 6 membered ring lactone, C|C8 alkyl group, C2 C8 alkenyl group, C2-Cs alkynyl group, aryl group, and glucosyl group Selective one or more than 8 201247612 is selected from nr5r6, 〇r5, oc(=o)r7, c(=〇)〇R5, c(=0)R5, C|-C8 alkyl, C2-C8 phenyl , C2-C8 block group, c3-C8 ring cyclyl, Ci-C8 dentate group, and C 丨-C8 pit oxygen substituent; each R5, and R6 are independently hydrogen, or CrC8 Alkyl; RACVCs alkyl, OR5, or NR5R6; m is 1 -12; and η is 1-12. Another embodiment of the present invention provides a method of inhibiting ROS/NO and/or p47ph°x of a subject comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof And a metabolite thereof, a solvate thereof, or a pre-drug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中,每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CpC^CVCs炫基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)R7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内酯、CVC8烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯< Ci-C8烷基、c2-c8 烯基' C^C:8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、 201247612 C,-C8院基、C2-C8稀基、(:2-<:8炔基、c3-C8環炫基、CVCs 鹵烧基、及C|-C8院氧基之取代基所取代; 每一 R_5、及R0係各自獨立為氫、或Ci_C8统基; 係 C,-C8烷基、〇r5、或NR5R6 ; m為1-12 ;以及 η為 1-12 。 本發明之另一實施態樣係在提供一種減少一主體之 CD3+/CD69+ Τ細胞之方法,包括:投予一有效劑量之一環 己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、其溶 劑化物、或其前趨藥物至該主體,其中,該環己烯酮化合 物係具有下列結構:Wherein each of the X and Y systems is independently oxygen, NR5, or sulfur; the R system is hydrogen, or a CpC^CVCs danic group; each R丨, R2, and R3 are each independently hydrogen, methyl, or ( CH2)m-CH3; R4 is NR5R6, 〇R5, 〇c(=〇)R7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, CVC8 alkyl, a C2-C8 alkenyl group, a C2-C8 alkynyl group, an aryl group, or a glucosyl group, wherein the 5 or 6 membered ring lactone < Ci-C8 alkyl group, c2-c8 alkenyl group C^C:8 alkynyl group, The aryl group and the glucosyl group are selectively selected from nr5r6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, 201247612 C,-C8, C2 -C8 dilute, (: 2-<:8 alkynyl, c3-C8 cyclodextrin, CVCs haloalkyl, and C|-C8 substituents substituted; each R_5, and R0 are each Independently hydrogen, or Ci_C8; C, -C8 alkyl, 〇r5, or NR5R6; m is 1-12; and η is 1-12. Another embodiment of the present invention provides a reduction of one A method for CD3+/CD69+ sputum cells of a subject, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solution thereof a compound, or a pre-drug thereof, to the host, wherein the cyclohexenone compound has the following structure:

其中,每一 X及Υ係各自獨立為氧、Nr5、或硫; R係為氫、或CbO^-Ce烷基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)r7、c(=〇)〇R5、C(=0)R5、鹵 素、5或6員環内醋、crC8燒基、c2-C8稀基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Ci_C8烷基、C2_C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、0R5、〇C(=〇)R7、c(=〇)〇R5、c(=〇)R5、 C「c8院基、c2-c8稀基 ' c2-C8块基、(:3-(:8環烧基、CrC8 鹵炫基、及C|-C8院氧基之取代基所取代; 201247612 每一 R5、及R0係各自獨立為氫、或Ci_Ce烷基; I係 crci{烧基、〇r5、或 nr5r6 ; m為1-12 ;以及 η為 1-12 。 本發明之另-實施態樣係在提供一種提升腎磁中穀耽 甘肽過氧化酶(GPx)活性之方法,包括:投予—有效劑量之 -環己烯,化合物、其醫藥上可接受之鹽類、其代謝物、 其溶劑化物、或其前趨藥物至該主體,其中,該環己烯酮 化合物係具有下列結構:Wherein each X and oxime is independently oxygen, Nr5, or sulfur; R is hydrogen or CbO^-Ce alkyl; each R丨, R2, and R3 are each independently hydrogen, methyl, or (CH2)m_CH3 ; R4 is NR5R6, 〇R5, 〇c(=〇)r7, c(=〇)〇R5, C(=0)R5, halogen, 5 or 6 member ring vinegar, crC8 alkyl, a c2-C8 dilute group, a C2-C8 alkynyl group, an aryl group, or a glucosyl group, wherein the 5 or 6 membered ring lactone, Ci_C8 alkyl group, C2_C8 alkenyl group, CrC8 alkynyl group, aryl group, and glucosyl group selectivity One or more selected from the group consisting of NR5R6, 0R5, 〇C(=〇)R7, c(=〇)〇R5, c(=〇)R5, C"c8 yard base, c2-c8 base' c2-C8 block Substituting (: 3-(:8-cycloalkyl), CrC8 halo-based, and C|-C8-substituted substituents; 201247612 Each R5, and R0 are each independently hydrogen or Ci_Ce alkyl; I system crci {alkyl, 〇r5, or nr5r6; m is 1-12; and η is 1-12. Another aspect of the present invention provides a glutathione peroxidase that enhances renal magnetic properties ( GPx) methods of activity comprising: administering an effective amount of cyclohexene, a compound, a pharmaceutically acceptable salt thereof, a metabolite, a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中,每一 X及Y係各自獨立為氧、nr5、或硫; R係為氫、或¢:(=0)(^-(:8烷基; 每一R丨、R2、及R3係各自獨立為氫、甲基 '或(CH2)m_CH3; R4係為 NR5R6、〇R5、〇C(=〇)R7、c(=〇)〇R5、c(=〇)R5、鹵 素、5或6員環内醋、炫基、C2-C8+缚基、C2-C8块基、 芳基、或葡萄糖基,其中,5或ό員環内酯、CrCs烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇c(=〇)R7、c(=0)0R5、c(=o)r5、 基、C2-C8稀基、c2-c8快基、c3-c8環炫基、CVCs 鹵院基、及CrCs烧氧基之取代基所取代; 每一尺5'及尺6係各自獨立為氩、或c「C8烧基; R7係 C「C8烧基、〇R5、4NR5R6 ; 201247612 m為1 -12 ;以及 η為 1-12。 本發明之另一實施態樣係在提供一種減少一主體之促 炎細胞因子之方法,包括:投予一有效劑量之一環己烯酮 化合物、其醫藥上可接受之鹽類、其代謝物、其溶劑化物、 或其前趨藥物至該主體,其中,該環己烯酮化合物係具有 下列結構:Wherein each X and Y system is independently oxygen, nr5, or sulfur; R is hydrogen, or ¢: (=0) (^-(:8 alkyl; each R丨, R2, and R3 are each Independently hydrogen, methyl 'or (CH2)m_CH3; R4 is NR5R6, 〇R5, 〇C(=〇)R7, c(=〇)〇R5, c(=〇)R5, halogen, 5 or 6 members Ring vinegar, leuco, C2-C8+, C2-C8 block, aryl, or glucosyl, wherein 5 or steroidal lactone, CrCs alkyl, C2-C8 alkenyl, CrC8 alkynyl, The aryl group and the glucosyl group are one or more selected from the group consisting of NR5R6, 〇R5, 〇c(=〇)R7, c(=0)0R5, c(=o)r5, yl, C2-C8 , c2-c8 fast radical, c3-c8 cyclodextrin, CVCs halogen-based, and CrCs alkoxy substituents; each 5' and 6-foot each independently argon, or c "C8 alkyl R7 is a C"C8 alkyl group, 〇R5, 4NR5R6; 201247612 m is 1 -12; and η is 1-12. Another embodiment of the present invention provides a method for reducing a proinflammatory cytokine of a subject. Including: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solvate thereof A pharmaceutical or a predecessor to the body, wherein the cyclohexenone compound had the following structure:

其中,每一X及Υ係各自獨立為氧、NR5 '或硫; R係為氫、或CbOA-Cs燒基; 每一Ri、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)R7、c(=o)or5、C(=0)R5、鹵 素、5或ό員環内酯、crC8烷基、C2-C8烯基、C2-C8炔基、 芳基'或葡萄糖基’其中,5或6員環内酯、CrC8烷基、C2-C8 稀基、C2_CS炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇c(=〇)r7、c(=o)or5、c(=o)r5、 CVCV^ 基、C2-C8烯基 ' c2-C8炔基 ' (:3-(:8環烷基、CVC8 商烧基、及Ci-C8烷氧基之取代基所取代; 每R5、及R6係各自獨立為氮、或Ci-Cg烧基; R7係C|-C8烷基、〇r5、或nr5r6 ; m為1-12 ;以及 η為 M2。 12 201247612 本發明之另一實施態樣係在提供一種減少腎臟中硫胱 胺酸蛋白酶-1表現及/或抑制腎NLRP3活性之方法,包括: 投予一有效劑量之一環己烯酮化合物、其醫藥上可接受之 鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主體, 其中,該環己烯酮化合物係具有下列結構:Wherein each X and oxime is independently oxygen, NR5' or sulfur; R is hydrogen or CbOA-Cs alkyl; each of Ri, R2, and R3 is independently hydrogen, methyl, or (CH2) m-CH3; R4 is NR5R6, 〇R5, 〇c(=〇)R7, c(=o)or5, C(=0)R5, halogen, 5 or steroidal lactone, crC8 alkyl, C2 -C8 alkenyl, C2-C8 alkynyl, aryl ' or glucosyl' wherein 5 or 6 membered ring lactones, CrC8 alkyl groups, C2-C8 dilute groups, C2_CS alkynyl groups, aryl groups, and glucosyl group selection One or more selected from the group consisting of NR5R6, 〇R5, 〇c(=〇)r7, c(=o)or5, c(=o)r5, CVCV^, C2-C8 alkenyl c2-C8 alkynyl '(: 3-(:8-cycloalkyl, CVC8 comonyl, and Ci-C8 alkoxy substituents are substituted; each R5, and R6 are independently nitrogen or Ci-Cg alkyl; R7 C|-C8 alkyl, 〇r5, or nr5r6; m is 1-12; and η is M2. 12 201247612 Another embodiment of the present invention provides a method for reducing thiocysteine-1 expression in the kidney and / or a method of inhibiting renal NLRP3 activity, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite, a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中,每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或C(=0)C「C8烷基; 每一 Ri、:R2、及R3係各自獨立為氫、曱基、或(CH2)m-CH3 ; R4係為 NR5R6、OR5、oc(=o)r7、C(=〇)〇R5、c(=〇)R5、函 素、5或6員環内酯、C〗-C8烧基、c2-c8烯基、C2-C8块基、 芳基、或葡萄糖基’其中,5或6員環内酯、〇^-(:8烷基、C2-C8 晞基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、0C(=0)R7、c(=o)or5、c(=o)r5、Wherein each of the X and Y systems is independently oxygen, NR5, or sulfur; R is hydrogen, or C(=0)C "C8 alkyl; each Ri, R2, and R3 are each independently hydrogen, Indenyl, or (CH2)m-CH3; R4 is NR5R6, OR5, oc(=o)r7, C(=〇)〇R5, c(=〇)R5, element, 5 or 6 membered ring lactone , C--C8 alkyl, c2-c8 alkenyl, C2-C8 block, aryl, or glucosyl' wherein 5 or 6 membered ring lactones, 〇^-(:8 alkyl, C2-C8 晞One or more selected from the group consisting of NR5R6, OR5, 0C(=0)R7, c(=o)or5, c(=o)r5,

Ci-Cg:^ 基、C2-C8稀基、C2-C8块基、C3-C8環炫•基、Cj-C8 鹵烷基、及c「c8烷氧基之取代基所取代; 每一R5、及116係各自獨立為氫、或Crc8烷基; R7係 Ci-Cg烧基、OR5、或NH ; m為1 · 12 ;以及 η 為 1-12。 本發明之再一實施態樣係在提供一種減少腎臟中腎 NF-κΒ量之方法,包括:投予一有效劑量之一環己烯酮化合 13 201247612 物、其醫藥上可接受之鹽類、其代謝物、其溶劑化物、或 其前趨藥物至該主體,其中,該環己烯酮化合物係具有下 列結構:Substituted by a substituent of Ci-Cg: C2-C8, C2-C8, C3-C8 cyclohexyl, Cj-C8 haloalkyl, and c"c8 alkoxy; each R5 And 116 are each independently hydrogen or a Crc8 alkyl group; R7 is a Ci-Cg alkyl group, OR5, or NH; m is 1 · 12; and η is 1-12. A further embodiment of the present invention is Provided is a method for reducing the amount of renal NF-κΒ in a kidney comprising: administering an effective dose of one of cyclohexenone compounds 13 201247612, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a precursor thereof The drug is applied to the host, wherein the cyclohexenone compound has the following structure:

其中,每一X及Y係各自獨立為氧、nr5、或硫; R係為氫、或(:(=0)<:|-(:8烷基; 每一 Ri、R2、及R3係各自獨立為氫、曱基、或(CH2)m-CH3 ; R4係為 NR5R6、0R5、〇c(=o)r_7、C(=0)0R5、C(=0)R5、齒 素、5或6員環内酯、C!-C8烧基、C2-C8稀基、c2-c8快基、 芳基、或葡萄糖基’其中,5或6員環内酯、C,-C8烷基、C2-C8 稀基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6、or5、〇c(=o)r7、c(=o)or5、c(=o)r5、 Ci-Cs院基、c2-c8稀基、c2-c8快基、c3-c8環炫基、Ci-Ce 齒烷基、及C丨-C8烷氧基之取代基所取代; 每一Rs、及Re係各自獨立為氫、或Ci-Cs烷基; R7係 cvc8烷基、or5、或NR5R6; m為1-12 ;以及 π 為 1 -12。 本發明之再一實施態樣係在提供一種抑制腎臟中細胞 凋亡之方法,包括:投予一有效劑量之一環己烯酮化合物、 其醫藥上可接受之鹽類、其代謝物、其溶劑化物、或其前 14 201247612 趨藥物至該主體,其中,該環己烯酮化合物係具有下列結 構:Wherein each X and Y system is independently oxygen, nr5, or sulfur; R is hydrogen, or (:(=0)<:|-(:8 alkyl; each Ri, R2, and R3 Each is independently hydrogen, sulfhydryl, or (CH2)m-CH3; R4 is NR5R6, 0R5, 〇c(=o)r_7, C(=0)0R5, C(=0)R5, dentate, 5 or 6-membered ring lactone, C!-C8 alkyl group, C2-C8 dilute group, c2-c8 fast group, aryl group, or glucosyl group] wherein 5 or 6 membered ring lactone, C, -C8 alkyl group, C2 -C8 dilute, CrC8 alkynyl, aryl, and glucosyl selective one or more selected from nr5r6, or5, 〇c(=o)r7, c(=o)or5, c(=o)r5 Substituting for a substituent of a Ci-Cs, a c2-c8 dilute group, a c2-c8 fast group, a c3-c8 cyclodyl group, a Ci-Ce adentate group, and a C丨-C8 alkoxy group; each Rs And Re are each independently hydrogen or a Ci-Cs alkyl group; R7 is a cvc8 alkyl group, or5, or NR5R6; m is 1-12; and π is 1 -12. Yet another embodiment of the present invention is Provided is a method for inhibiting apoptosis in a kidney, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a precursor thereof 4 201247612 Drugs to the host, wherein the cyclohexenone compound has the following structure:

其中,每一X及Y係各自獨立為氧、NR5、或硫; R係為氫,‘或C(=〇)Cl-C8烧基; 每一R,、r2、及r3係各自獨立為氫、曱基、或(CH2)m_CH3 ; R4係為 NR5R6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、卤 素、5或6員環内酯、C,-C8烷基、c2-c8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中’ 5或6員環内酯、CrC8烷基、C2-C8 稀基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6、OR5、0C(=0)R7、c(=o)or5、c(=o)r5、 CrC8院基、(:2-0:8稀基、c2-c8快基、C3-C83t烧基、CVC8 i烷基、及CrCs烷氧基之取代基所取代; 每一 Rs、及R6係各自獨立為氫、或C^-Cs烷基; R7係 Crc8烷基、or5、或 nr5r6 ; m為1-12 :以及 η為 1-12 。 本發明之另一實施態樣係在提供一種保護或預防一主 體腎臟罹患腎絲球硬化症及/或腎絲球腎炎之方法,包括: 投予一有效劑量之一環己烯酮化合物、其醫藥上可接受之 鹽類、其代謝物、其溶劑化物 '或其前趨藥物至該主體, 15 201247612 以減少腎臟中TGHM蛋白之表現程度及_蛋白丨、眼ιν 蛋白聚集’其中’該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, NR5, or sulfur; R is hydrogen, or C (=〇)Cl-C8 alkyl; each R, r2, and r3 are each independently hydrogen , fluorenyl, or (CH2)m_CH3; R4 is NR5R6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, C, -C8 alkyl, c2-c8 alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein '5 or 6 membered ring lactone, CrC8 alkyl, C2-C8 dilute, CrC8 alkynyl, aryl And one or more selected from the group consisting of nr5r6, OR5, 0C(=0)R7, c(=o)or5, c(=o)r5, CrC8, and (:2-0:8) Substituted by a substituent of a dilute group, a c2-c8 fast group, a C3-C83t alkyl group, a CVC8 i alkyl group, and a CrCs alkoxy group; each of the Rs and R6 groups is independently hydrogen or a C^-Cs alkyl group R7 is Crc8 alkyl, or5, or nr5r6; m is 1-12: and η is 1-12. Another embodiment of the present invention provides protection or prevention of renal sclerosing in a subject kidney / or method of glomerulonephritis, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, Metabolite, solvate 'or the precursor of the drug to the body, in order to reduce the kidney 15201247612 TGHM protein and levels of expression of protein _ Shu, eye ιν protein aggregation' where 'the cyclohexenone compound had the following structure:

其中,每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CpC^CVCs烷基; 每一 R,、R2、及R3係各自獨立為氫、甲基、或(CH2)m CH3 ; R4係為 NR5R6、OR5、〇C(=〇)R7、c(=〇)OR5、c卜〇)r5、卤 素、5或6員環内醋、CVCs烧基、(:2-0:8稀基、c2-c8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Cl_C8烷基、C2_C8 烯基、C2_C:8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、〇c(=〇)r7、c(=〇)〇R5、c(=〇)R5、 c】-c8烧基、c2-c8烯基' c2-c8块基、C3_C8環烧基、C|_Cg 鹵烧基、及Ci-Cg院氧基之取代基所取代; 每一Rs、及係各自獨立為氫、或C|-C8烷基; R7係Ci-Cg烧基、OR5、或NR5R6 ; m為1-12 ;以及 η為1-12 。 本發明之再一實施態樣係在提供一種治療一主體之局 部性腎絲球硬化症(FSGS)之方法,包括:投予一有效劑量 之一環己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、 其溶劑化物、或其前趨藥物至該主體,以(i)增加腎臟中 Nrf2活性及/或(ii)抑制腎臟中NF-κΒ相關發炎反應及 201247612 TGF-βΙ誘導之纖維化’其中,該環己烯酮化合物係具有下 列結構:Wherein each of the X and Y systems is independently oxygen, NR5, or sulfur; R is hydrogen, or CpC^CVCs alkyl; each R, R2, and R3 are each independently hydrogen, methyl, or ( CH2)m CH3 ; R4 is NR5R6, OR5, 〇C(=〇)R7, c(=〇)OR5, c 〇)r5, halogen, 5 or 6 member ring vinegar, CVCs alkyl, (: 2 -0:8 dilute, c2-c8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, Cl_C8 alkyl, C2_C8 alkenyl, C2_C:8 alkynyl, aryl, and glucosyl Selectively one or more selected from the group consisting of NR5R6, OR5, 〇c(=〇)r7, c(=〇)〇R5, c(=〇)R5, c]-c8 alkyl, c2-c8 alkenyl' Substituting a c2-c8 block group, a C3_C8 cycloalkyl group, a C|_Cg haloalkyl group, and a substituent of a Ci-Cg alkoxy group; each Rs, and each of the groups is independently hydrogen or a C|-C8 alkyl group; R7 is a Ci-Cg alkyl group, OR5, or NR5R6; m is 1-12; and η is 1-12. A further aspect of the present invention provides a method for treating a subject with localized squamous sclerosis ( The method of FSGS) comprises: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, and metabolism thereof a solvate thereof, or a pre-drug thereof, to (i) increase Nrf2 activity in the kidney and/or (ii) inhibit NF-κΒ-related inflammatory response in the kidney and 201247612 TGF-βΙ-induced fibrosis, wherein The cyclohexenone compound has the following structure:

其中’每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫,或CpCOCVCe烷基; 每一R!、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NEL5R6、or5、oc(=o)r7、c(=o)or5、C(=〇)R5、鹵 素、5或6員環内酯、CVCe烷基、c2-c8烯基、c2-c8炔基、 芳基、或琦萄糖基,其中,5或6員環内酯、CrCs烷基、c2-C8 稀基、Cr-C8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6、OR5、〇C(=〇)R7、C(=0)OR5、c(=〇)r5、 CrC8燒基、(:2-(:8稀基、C2-C8炔基、C3-C8環烧基、CVCs 齒烧基、及C丨-C8烷氧基之取代基所取代; 每一R·5、及R·6係各自獨立為氫、或c丨-c8烷基; R?係 cvc8烷基、〇r5、或nr5r6 ; m為1 -12 以及 η為 1-12 。 本發明之再一.實施態樣係在提供一種治療一主體之腎 絲球腎炎之方法,包括:投予一有效劑量之一環己烯酮化 合物、其醫藥上可接受之鹽類、其代謝物、其溶劑化物、 或其前趨藥物至該主體,以(〇抑制腎NLRp3發炎體活化及 17 201247612 /或(11)抑制T細胞活性增加,其中,該環己烯酮化合物係 具有下列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or CpCOCVCe alkyl; each R!, R2, and R3 are each independently hydrogen, methyl, or (CH2) m_CH3 ; R4 is NEL5R6, or5, oc(=o)r7, c(=o)or5, C(=〇)R5, halogen, 5 or 6 membered ring lactone, CVCe alkyl, c2-c8 alkenyl, a c2-c8 alkynyl, aryl, or glucosyl group, wherein 5 or 6 membered ring lactones, CrCs alkyl groups, c2-C8 dilute groups, Cr-C8 alkynyl groups, aryl groups, and glucosyl group selectivity One or more selected from nr5r6, OR5, 〇C(=〇)R7, C(=0)OR5, c(=〇)r5, CrC8 alkyl, (:2-(:8 dilute, C2-C8) Substituted by alkynyl, C3-C8 cycloalkyl, CVCs dentate, and C丨-C8 alkoxy substituents; each R·5, and R·6 are each independently hydrogen or c丨-c8 An alkyl group; R? is a cvc8 alkyl group, 〇r5, or nr5r6; m is 1 -12 and η is 1-12. A further aspect of the present invention provides a method for treating a subject of renal glomerulonephritis The method comprises: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, Or a pre-existing drug to the host to inhibit the activation of renal NLRp3 inflammatory body and 17 201247612 / or (11) to inhibit the increase of T cell activity, wherein the cyclohexenone compound has the following structure:

其中’每一X及γ係各自獨立為氧、NR5、或硫; R係為氫、或CpcOCi-Cg烷基; 每一Ri、R2、及r3係各自獨立為氬、曱基、或(CH2)m_CH3 ; R4係為 NhR6、or5、oc(=o)r7 ' c(=o)or5、c(=o)r5、鹵 素、5或6員環内醋、c丨·c8烧基、c2_c8稀基、C2_C8炔基、 芳基、或葡萄糖基,其中’5或6員環内酯、Ci-Cs烷基、C2-C8 稀基、CyC:8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6、or5、〇c(=o)r7、c(=o)or5、c(=o)r5、 Ci-C8烧基、c2-C8稀基、C2-C8炔基、C3-C8環炫《基、Ci-Cs i炫基、及crc8烷氧基之取代基所取代; 每一、及心係各自獨立為氫、或C丨_C8烷基; R_7係Ci-C8烧基、〇R5、或nr5r6 ; m為1-12 ;以及 η為 1-12 。 本發明之再一實施態樣係在提供一種減缓一主體之免 疫球蛋白Α型腎絲球腎炎(IgAN)之方法,包括:投予一有效 劑量之一環己烯酮化合物、其醫藥上可接受之鹽類、其代 謝物、其溶劑化物、或其前趨藥物至該主體,以(丨)提升Nrf2 18 201247612 活性及/或(ii)抑制腎臟中NF-κΒ相關發炎反應及TGF-βΙ誘 導之纖維化’其中’該環己烯酮化合物係具有下列結構:Wherein 'each X and γ are each independently oxygen, NR5, or sulfur; R is hydrogen, or CpcOCi-Cg alkyl; each Ri, R2, and r3 are independently argon, sulfhydryl, or (CH2) m_CH3; R4 is NhR6, or5, oc(=o)r7 'c(=o)or5, c(=o)r5, halogen, 5 or 6 member ring vinegar, c丨·c8 alkyl, c2_c8 a C2_C8 alkynyl, aryl, or glucosyl group, wherein '5 or 6 membered ring lactone, Ci-Cs alkyl, C2-C8 dilute, CyC:8 alkynyl, aryl, and glucosyl selective One or more selected from nr5r6, or5, 〇c(=o)r7, c(=o)or5, c(=o)r5, Ci-C8 alkyl, c2-C8 dilute, C2-C8 alkynyl , C3-C8 cyclodextrin, "substituent, Ci-Cs i dyl, and crc8 alkoxy substituents; each and each of the cores are independently hydrogen, or C丨_C8 alkyl; R_7 is Ci- C8 alkyl, hydrazine R5, or nr5r6; m is 1-12; and η is 1-12. A further aspect of the present invention provides a method for slowing down a subject of immunoglobulin-type glomerulonephritis (IgAN), comprising: administering an effective dose of one of cyclohexenone compounds, which is pharmaceutically acceptable Accepting a salt, a metabolite thereof, a solvate thereof, or a pre-drug thereof to the subject, (提升) enhancing Nrf2 18 201247612 activity and/or (ii) inhibiting NF-κΒ-related inflammatory response and TGF-βΙ in the kidney Induced fibrosis 'where' the cyclohexenone compound has the following structure:

其中’每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CpOK^-Cs烷基; 每一 R,、:R2、及R3係各自獨立為氫、曱基、或(CH2)m_CH3 ; R4係為 NR5R6 ' or5、oc(=o)r7、c(=o)or5、C(=0)R5、鹵 素、5或6員環内酯、(VC8烧基、c2-c8烯基、c2-c8快基、 芳基、或葡萄糖基,其中’5或6員環内酯、C「C8烷基、c2-C8 稀基、C2-C8快基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、0C(=0)R7、C(=0)〇R5、(^=〇)r5、 基、C2-C8烯基、C2-C8块基、c3-c8環貌基、Ci-Cs 鹵烷基、及C丨-C8烧氧基之取代基所取代; 每一Rs、及116係各自獨立為氫、或C,-C8烷基; R7係 C|-C8烷基、OR5、或NR5R6 ; m為1-12 ;以及 η 為 1 -12 〇 本發明所提到之所有文獻、專利、申請中專利之内容 均併入本發明之内容以供參考,也就是於本發明中明確且 分別提到的各個文獻、專利、或申請中專利均併入本發明 以供參考。 201247612 腎絲球疾病包括多種基因及環境因素所導致之許多病 症,但不外乎主要分成腎絲球硬化症及腎絲球腎炎兩大 類。腎絲球硬化症,特別是FSGS,其主要應該是起因於腎 絲球上皮細胞導致腎絲球中複雜的反應產生。這些複雜的 反應可包括氧化壓力、巨噬細胞聚集之發炎反應、及促進 基質產生及/或基質分解之因子。腎踺中過多的活化系統性 T細胞及淋巴球/巨噬細胞/嗜中性白血球浸潤,容易造成免 疫球蛋白A型(IgAN)加速發展,此為常見的源發性腎絲球腎 炎。然而’至今無論是IgAN的聚集及惡化程度之預防及治 療上’仍未大量的受到研究。在此,本發明提供一種治療 腎病的方法,特別是治療腎絲球硬化症或腎絲球腎炎之方 法,其係透過投予本發明所提供之環己烯酮化合物至一主 體(如’人類)》此環己烯酮化合物對一主體(特別是腎職)具 有治療效果’而可用以治療腎絲球硬化症(如實施例16、14 所示)及/或腎絲球腎炎(如實施例7-13、14所示)》 於部分實施例中’係提供一種治療一主艘之腎病(如腎 絲球硬化症或腎絲球腎炎)的方法。此方法包括:投予—有 效劑量之一環己烯酮化合物、其醫藥上可接受之鹽類其 代謝物、其溶劑化物、或其前趨藥物至該主體,其中,該 環己烯酮化合物係具有下列結構: ΛWherein each of X and Y is independently oxygen, NR5, or sulfur; R is hydrogen or CpOK^-Cs alkyl; each R,, R2, and R3 are independently hydrogen, sulfhydryl, Or (CH2)m_CH3; R4 is NR5R6 ' or5, oc(=o)r7, c(=o)or5, C(=0)R5, halogen, 5 or 6 membered ring lactone, (VC8 alkyl, c2 a -c8 alkenyl group, a c2-c8 fast group, an aryl group, or a glucosyl group, wherein '5 or 6 membered ring lactones, C "C8 alkyl group, c2-C8 dilute group, C2-C8 fast group, aryl group, and One or more selected from the group consisting of NR5R6, OR5, 0C(=0)R7, C(=0)〇R5, (^=〇)r5, phenyl, C2-C8 alkenyl, C2-C8 Substituted by a substituent of a c3-c8 ring group, a Ci-Cs haloalkyl group, and a C丨-C8 alkoxy group; each Rs and 116 groups are each independently hydrogen or a C,-C8 alkyl group; R7 is C|-C8 alkyl, OR5, or NR5R6; m is 1-12; and η is 1 -12. All the documents, patents, and patents mentioned in the present invention are incorporated in the contents of the present invention. For reference, that is, the various documents, patents, or patents that are expressly incorporated herein by reference in their entirety are hereby incorporated by reference. 201247612 Kidney spheroid disease includes many diseases caused by a variety of genes and environmental factors, but it is mainly divided into two categories: glomerulosclerosis and glomerulonephritis. Kidney sclerosing, especially FSGS, should be mainly The spheroidal epithelial cells are responsible for the complex reactions in the glomerulus. These complex reactions can include oxidative stress, inflammatory responses to macrophage aggregation, and factors that promote matrix production and/or matrix breakdown. Activation of systemic T cells and lymphocytic/macrophage/neutrophil infiltration, easily lead to the accelerated development of immunoglobulin A (IgAN), which is a common source of glomerulonephritis. However, The prevention and treatment of the degree of aggregation and deterioration of IgAN has not been extensively studied. Here, the present invention provides a method for treating kidney disease, particularly a method for treating glomerulosclerosis or glomerulonephritis, which is The cyclohexenone compound provided by the present invention is administered to a host (e.g., 'human). The cyclohexenone compound has a subject (especially a kidney) The therapeutic effect can be used to treat renal sclerosing (as shown in Examples 16, 14) and/or glomerulonephritis (as shown in Examples 7-13, 14). A method for treating a kidney disease of a main vessel, such as glomerulosclerosis or glomerulonephritis. The method comprises: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, and a metabolite thereof And a solvate thereof, or a pre-drug thereof, to the host, wherein the cyclohexenone compound has the following structure: Λ

其中’每一X及Υ係各自獨立為氧、NR5、或硫; 20 201247612 R係為氫、或<:(=0)(^-(:8烷基; 每一 R,、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 NR5R6、OR5、〇C(=〇)R7、C(=0)0R5、c(=o)r5、鹵 素、5或6員環内酯、CrC8烷基、c2-C8烯基、c2-c8炔基、 芳基、或葡萄糖基,其中’5或6員環内酯、CrC8烧基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、OC(=〇)R7、C(=0)0R5、c(=o)r5、 CrCs烷基、c2-c8烯基、c2-c8炔基、C3-C8環烷基、c丨-c8 卤烧基、及C丨-C8烧氧基之取代基所取代; 每一Rs、及R6係各自獨立為氫、或(:丨-(:8烷基; R7係 Ci-Cg烧基、OR5、或NR5R6 ; m為1-12 ;以及 η為 1-12 〇 於部分實施例中,係提供一種治療腎絲球硬化症之方 法。於特定實施例中,腎絲球硬化症係為局部性腎絲球硬 化症(FSGS)或結節性腎絲球硬化症。於特定實施例中腎 絲球硬化症係為局部性腎絲球硬化症(FSGS)。於部分實施 例中,環己烯酮化合物可防止氧化壓力。於特定實施例中, 係透過降低TGF-βΙ及胞外基質蛋白質表現,以防止氧化壓 力。於部分實施例中,主體係為人類。於特定實施例十, 係透過增加腎細聦核因數_E2相關因數2 (Nrf2)活性以減少 氧化壓力》 ' 於部分實施例中’係提供一種治療腎絲球腎炎之方 法’特定實施例中,腎絲球腎炎係為免疫球蛋白八型腎絲 201247612 球腎炎(IgAN)。於部分實施例中,環己烯酮化合物可減少 主體之CD3+/CD69+ T細胞。於特定實施例中,環己烯酮化 合物可減少主體中促炎細胞因子。於特定實施例中,促炎 細胞因子包括MCP-1、IL-6、IL-Ιβ、IL-18、或其組合。於 部分實施例中,主體係為人類。 於部分實施例中,具有Wherein 'each X and oxime are each independently oxygen, NR5, or sulfur; 20 201247612 R is hydrogen, or <:(=0)(^-(:8 alkyl; each R, R2, and R3 is independently hydrogen, methyl or (CH2)m-CH3; R4 is NR5R6, OR5, 〇C(=〇)R7, C(=0)0R5, c(=o)r5, halogen, 5 Or 6-membered ring lactone, CrC8 alkyl, c2-C8 alkenyl, c2-c8 alkynyl, aryl, or glucosyl, wherein '5 or 6 membered ring lactone, CrC8 alkyl, C2-C8 alkenyl, The CrC8 alkynyl, aryl, and glucosyl group are selected from one or more selected from the group consisting of NR5R6, OR5, OC(=〇)R7, C(=0)0R5, c(=o)r5, CrCs alkyl, c2 Substituted by a substituent of -c8 alkenyl, c2-c8 alkynyl, C3-C8 cycloalkyl, c丨-c8 haloalkyl, and C丨-C8 alkoxy; each Rs, and R6 are independently Hydrogen, or (: 丨-(:8 alkyl; R7-based Ci-Cg alkyl, OR5, or NR5R6; m is 1-12; and η is 1-12 〇 in some embodiments, provides a therapeutic kidney A method of spheroid sclerosis. In a specific embodiment, the glomerulosclerosis is local glomerulosclerosis (FSGS) or nodular glomerulosclerosis. In the case of glomerulosclerosis, local glomerulosclerosis (FSGS) is used. In some embodiments, the cyclohexenone compound prevents oxidative stress. In a particular embodiment, the TGF-β Ι cell is reduced by transmission. The outer matrix protein is expressed to prevent oxidative stress. In some embodiments, the primary system is human. In a specific embodiment, the oxidative stress is reduced by increasing the renal factor _E2 correlation factor 2 (Nrf2) activity. In some embodiments, 'providing a method for treating glomerulonephritis', in a specific embodiment, the glomerulonephritis is an immunoglobulin type 8 kidney filament 201247612 glomerulonephritis (IgAN). In some embodiments, The enone compound reduces CD3+/CD69+ T cells in the subject. In a particular embodiment, the cyclohexenone compound reduces proinflammatory cytokines in the subject. In a particular embodiment, the proinflammatory cytokines include MCP-1, IL- 6. IL-Ιβ, IL-18, or a combination thereof. In some embodiments, the primary system is a human. In some embodiments,

結構之環己稀 酮化合物可由任何適當的起始材料經合成或半合成方式製 備。於其他實施例中,環己烯酮化合物可透過發酵作用、 或其他相似方法製備。例如,在某些情況下,由化合物1(即 熟知之安卓奎諾爾(Antroquinonol®或”Antroq”)、或化合物3 可從4-羥基-2,3-二曱氧基-6-甲基環己-2,5-二烯酮 (4-hydroxy-2,3-dimethoxy-6-methylcyclohexa-2,5-dienone) 所製備而得。其中,化合物之非限定例子如下所示。 ch3 ch3 ch3 ch3The cyclohexanone compound of the structure can be prepared synthetically or semi-synthetically from any suitable starting material. In other embodiments, the cyclohexenone compound can be prepared by fermentation, or other similar methods. For example, in some cases, compound 1 (known as Antroquinonol® or "Antroq"), or compound 3 may be derived from 4-hydroxy-2,3-dimethoxy-6-methyl ring Prepared by 4-hydroxy-2,3-dimethoxy-6-methylcyclohexa-2,5-dienone. Non-limiting examples of the compounds are shown below. ch3 ch3 ch3 ch3

0、 1 ch3 ch3 ch3 ch3 ch30, 1 ch3 ch3 ch3 ch3 ch3

22 20124761222 201247612

23 20124761223 201247612

OHOH

CH, CH-, CH, CH,CH, CH-, CH, CH,

結構之環己烯 酮化合物係為牛樟芝之有機溶劑萃取物。於部分實施例 24 201247612 中,有機溶劑係選自由醇類(如,甲醇、乙醇、丙醇、或豆 相似物)、醋类員(如’乙酸甲醋、乙酸乙醋、或其相似物)、 烧類(如’戊院、己烧、庚院、或其相似物)、_(如, 氯甲烷、氣乙垸、氣仿、二氣甲炫、或其相似物)、及豆相 =物。例如’示範化合物N7可從有機溶劑萃取物中分離而 得。於特定實施例令,有機溶劑係為醇類;且於特定實施 例中,此醇類係為乙醇。於部分實施例中,環己稀綱化合 物係由牛樟芝之水萃取物分離而得。 於部分實施例中,本發明之減緩腎功能損害或腎絲球 損傷之方法,包括:投予一有效劑量之一環己烯酮化合物、 其醫藥上可接受之鹽類、其代謝物、其溶劑化物、或其前 趨藥物至一主體,其中,該環己烯酮化合物係具有下列結 構: 'The cyclohexenone compound of the structure is an organic solvent extract of Antrodia camphorata. In some embodiments 24 201247612, the organic solvent is selected from the group consisting of alcohols (eg, methanol, ethanol, propanol, or bean analogs), vinegar members (eg, 'methyl acetate, ethyl acetate, or the like) , burning (such as 'Eight Court, burned, Gengyuan, or similar things), _ (eg, methyl chloride, gas, sputum, gas, two gas, or similar), and bean phase = Things. For example, the exemplary compound N7 can be isolated from an organic solvent extract. In a particular embodiment, the organic solvent is an alcohol; and in a particular embodiment, the alcohol is ethanol. In some embodiments, the cycloheximide compound is isolated from an aqueous extract of Antrodia camphorata. In some embodiments, the method of the invention for alleviating renal dysfunction or glomerular damage comprises: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solvent thereof. a compound, or a pre-drug thereof, to a host, wherein the cyclohexenone compound has the following structure:

其中’每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或c〇=o)crc8烷基; 每一Ri、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、〇R5 ' 〇c(=〇)R7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内酯、CrCs烷基' c2-c8烯基、C2-C8炔基、 芳基、或葡萄糖基’其中,5或6員環内酯、Ci-Cs烷基、c2-c8 稀基、Ci-Cg炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇c(=〇)R7、c(=o)or5、c(=o)r5、 25 201247612 C,-C8:^ 基、c2_C8 稀基、C2_C8 快基、C3_C4烧基、C|_C8 齒统基、及c「c8烷氡基之取代基所取代; 每一 R5、及R0係各自獨立為氬、或crc8烷基; R7係C,-C8烷基、〇r5、或Nr5r6 ; m為1 -12 ;以及 η為 1-12 。 於部分實施例中,腎絲球損傷包括上皮增生損傷 (EPHL) 〇於部分實施例中,主體係為人類。 於部分實施例中’本發明所提供之環己烯酮化合物具 有增加腎細胞核因數_Ε2相關因數2 (Nrf2)活性,但卻可抑 制腎贜中NF-κΒ相關發炎反應及TGF-βΙ誘導之纖維化路徑 之治療功效。請參見實施例5、6及14。 本發明之部分實施例係提供一種提升一主體之腎細胞 核因數-E2相關因數2 (Nrf2)活性之方法’包括投予一有效 劑量之一環己烯酮化合物、其醫藥上可接受之鹽類、其代 謝物、其溶劑化物、或其前趨藥物至一主趙,其中,該環 己烯酮化合物係具有下列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or c〇=o) crc8 alkyl; each of Ri, R2, and R3 is independently hydrogen, methyl, Or (CH2)m_CH3; R4 is NR5R6, 〇R5 ' 〇c(=〇)R7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, CrCs alkyl' C2-c8 alkenyl, C2-C8 alkynyl, aryl, or glucosyl' wherein 5 or 6 membered ring lactone, Ci-Cs alkyl, c2-c8 dilute, Ci-Cg alkynyl, aryl, And one or more selected from the group consisting of NR5R6, 〇R5, 〇c(=〇)R7, c(=o)or5, c(=o)r5, 25 201247612 C,-C8:^, C2_C8 dilute group, C2_C8 fast group, C3_C4 alkyl group, C|_C8 dentate group, and c"c8 alkyl fluorenyl substituent; each R5, and R0 are independently argon or crc8 alkyl; R7 C, -C8 alkyl, 〇r5, or Nr5r6; m is 1 -12; and η is 1-12. In some embodiments, renal glomerular damage includes epithelial hyperplasia (EPHL), in some embodiments The main system is human. In some embodiments, the cyclohexenone compound provided by the present invention has an increase in renal cell nuclear factor. _Ε2 correlation factor 2 (Nrf2) activity, but inhibits the NF-κΒ-related inflammatory response in the renal pelvis and the therapeutic efficacy of the TGF-βΙ-induced fibrotic pathway. See Examples 5, 6 and 14. Partial implementation of the present invention A method for increasing the renal cell nuclear factor-E2 correlation factor 2 (Nrf2) activity of a subject comprises administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solvate, or a prodrug thereof, to the main compound, wherein the cyclohexenone compound has the following structure:

其中’每一 X及γ係各自獨立為氧、Nr5、或硫; R係為氫、或C(=0)C,-C8烷基; 每一 R〇、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; 26 201247612 R4係為 NR5R6、OR5、〇c(=〇)r7、C(=〇)〇R5、C(=0)R5、鹵 素、5或6員環内醋、CrC8炫基、c2-C8烯基、C2-C8快基、 芳基、或葡萄糖基’其中,5或6員環内酯、C「C8烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5' 〇c(=〇)r7、C(=0)0R5、C(=0)R5、 基、c2-c8稀基、C2-C8快基、C3-C8環炫《基、CVCs 齒炫基、及C「C8炫氧基之取代基所取代; 每一Rs、及R6係各自獨立為氫、或C1_C8烷基;Wherein 'each X and γ are each independently oxygen, Nr5, or sulfur; R is hydrogen, or C(=0)C, -C8 alkyl; each R〇, R2, and R3 are each independently hydrogen , methyl, or (CH2)m_CH3 ; 26 201247612 R4 is NR5R6, OR5, 〇c(=〇)r7, C(=〇)〇R5, C(=0)R5, halogen, 5 or 6 member ring Vinegar, CrC8 leucoyl, c2-C8 alkenyl, C2-C8 fast radical, aryl, or glucosyl' wherein 5 or 6 membered cyclic lactones, C"C8 alkyl, C2-C8 alkenyl, CrC8 alkynyl , one or more selected from the group consisting of NR5R6, 〇R5' 〇c(=〇)r7, C(=0)0R5, C(=0)R5, yl, c2-c8 Substituent, C2-C8 fast radical, C3-C8 cyclosporin "substituent, CVCs dentate, and C" C8 methoxyl substituent; each Rs, and R6 are each independently hydrogen, or C1_C8 alkyl ;

RySCVCs烧基、〇R5、或 NR5R6 ; m為1-12 ;以及 η為 1-12 。 於部分實施例中,主體係為人類。 本發明之部分實施例係提供一種抑制一主鱧之腎 NF-kB活性及/或轉化生長因子蛋白表現之方法, 包括:投予一有效劑量之一環己烯酮化合物、其醫藥上可 接受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至一 主體,其中,該環己烯酮化合物係具有下列結構:RySCVCs alkyl, hydrazine R5, or NR5R6; m is 1-12; and η is 1-12. In some embodiments, the primary system is a human. Some embodiments of the present invention provide a method of inhibiting renal NF-kB activity and/or transforming growth factor protein expression of a host, comprising: administering an effective dose of one of a cyclohexenone compound, which is pharmaceutically acceptable a salt, a metabolite thereof, a solvate thereof, or a prodrug thereof to a host, wherein the cyclohexenone compound has the following structure:

其中’每一 X及Y係各自獨立為氧、Nr5、或硫; R係為氫、或C(=〇)C,-C8烷基;Wherein each X and Y system is independently oxygen, Nr5, or sulfur; R is hydrogen, or C(=〇)C, -C8 alkyl;

每R|、R2、及R3係各自獨立為氫、甲基 '或(CH2)m_CH 27 201247612 R4係為 NR5R6、OR5、oc(=o)r7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内酯、Ci-Cs烷基、C2-C8烯基、c2-c8炔基、 芳基、或葡萄糖基’其中,5或6員環内醋、Ci-Cg炫基、C2-C8 烯基、C2-C8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、0C(=0)R7、C(=0)0R5、C(=0)R5、 Ci-Cg烧基、C2.Cs稀基、Cz-Cg快基、C3-C8環烧基、Ci-Cg 卤烷基、及crc8烷氧基之取代基所取代; 每一R5、及R6係各自獨立為氩、或C,-C8烷基; R7係C|-C8烷基、OR5、或NR5R6 ; m為1-12 ;以及 η為 1-12 。 於部分實施例中,主體係為人類。 於部分實施例中,當投予本發明所提供之環己烯酮化 合物(如.化合物1),可抑制腎臟中r〇s/n〇及p47ph〇x NAD(P)H氧化酶產生,但明顯的增加在FSGS主趙上與環己 烯酮化合物作用相關之Nrf2訊息傳導路徑。請參見實施例 3、5及 14。 本發明之部分實施例係提供一種抑制一主體之 ROS/NO及/或p47~之方法,包括:投予一有效劑量之一環 己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、其溶 劑化物、或其前趨藥物至該主體,其中,該環己烯嗣化合 物係具有下列結構: 28 201247612Each R|, R2, and R3 is independently hydrogen, methyl' or (CH2)m_CH 27 201247612 R4 is NR5R6, OR5, oc(=o)r7, c(=o)or5, c(=o) R5, halogen, 5 or 6 membered ring lactone, Ci-Cs alkyl, C2-C8 alkenyl, c2-c8 alkynyl, aryl, or glucosyl' wherein 5 or 6 membered vinegar, Ci-Cg One or more selected from the group consisting of NR5R6, 〇R5, 0C(=0)R7, C(=0)0R5, thiol, C2-C8 alkenyl, C2-C8 alkynyl, aryl, and glucosyl. Substituted by a substituent of C(=0)R5, Ci-Cg alkyl, C2.Cs, Cz-Cg, C3-C8 cycloalkyl, Ci-Cg haloalkyl, and crc8 alkoxy; Each of R5 and R6 is independently argon or C,-C8 alkyl; R7 is C|-C8 alkyl, OR5, or NR5R6; m is 1-12; and η is 1-12. In some embodiments, the primary system is a human. In some embodiments, administration of the cyclohexenone compound (eg, Compound 1) provided by the present invention inhibits the production of r〇s/n〇 and p47ph〇x NAD(P)H oxidase in the kidney, but Significantly increase the Nrf2 signaling pathway associated with the action of cyclohexenone compounds on FSGS. See Examples 3, 5, and 14. Some embodiments of the present invention provide a method of inhibiting ROS/NO and/or p47~ of a subject, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, and a metabolite thereof And a solvate thereof, or a prodrug thereof, to the host, wherein the cyclohexene oxime compound has the following structure: 28 201247612

其中’每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或(:(=0)0^-(:8烷基; 每一 R!、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、OR5、OC(=〇)R7、c(=〇)〇r5、c(=0)R5、鹵 素、5或6員環内醋、Cl-C8燒基、c2_C8稀基、C2_c8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、CrC8烷基、C2-C8 烯基、Cz-C:8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、〇C(=〇)r7、c(=〇)〇R5、c(=o)r5、 •基、C2-C8烯基、c2-c8炔基、c3-C8環炫•基、c,-c8 鹵炫基、及Ci-Cs烧氧基之取代基所取代; 每一 R5、及R6係各自獨立為氫、或Cl_c8烷基; R>7係 C!-Cg烧基、OR5、或NR5R6 ; m為1-12 ;以及 η 為 1-12。 於部分實施例中,主體係為人類。 本發明之部分實施例係提供一種減少一主趙之 CD3+/CD69+ Τ細胞之方法,包括:投予一有效劑量之一環 己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、其溶 劑化物、或其前趨藥物至該主體,其中,該環己烯酮化合 物係具有下列結構: 29 201247612Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or (:(=0)0^-(:8 alkyl; each R!, R2, and R3 are each Independently hydrogen, methyl, or (CH2)m_CH3; R4 is NR5R6, OR5, OC(=〇)R7, c(=〇)〇r5, c(=0)R5, halogen, 5 or 6 membered ring Vinegar, Cl-C8 alkyl, c2_C8 dilute, C2_c8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, CrC8 alkyl, C2-C8 alkenyl, Cz-C:8 alkynyl One or more selected from the group consisting of NR5R6, OR5, 〇C(=〇)r7, c(=〇)〇R5, c(=o)r5, • base, C2-C8 Alkenyl, c2-c8 alkynyl, c3-C8 cyclohexanyl, c,-c8 halo, and Ci-Cs alkoxy substituents; each R5, and R6 are independently hydrogen, Or Cl_c8 alkyl; R>7 is C!-Cg alkyl, OR5, or NR5R6; m is 1-12; and η is 1-12. In some embodiments, the main system is human. The invention provides a method for reducing a CD3+/CD69+ Τ cell of a main Zhao, comprising: administering an effective dose of one cyclohexenone compound, which is pharmaceutically acceptable Salts, their metabolites, solvates thereof, pharmaceutical or a predecessor to the body, wherein the cyclohexenone-based compound having the following structure: 29 201 247 612

其中,每一X及γ係各自獨立為氧、Nr5、或硫; R係為氫、或¢:(=0)(:,-(:8烧基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)r7、c(=〇)OR5、c卜〇)r5、鹵 素、5或6員環内酯、C|-C8烧基、c2-C8稀基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Ci_c8烷基、c2_C8 烯基、炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇C(=〇)R7、c(=0)OR5、c(=o)r5、 C「c8统基、c2-c8稀基、c2-c8块基、C3-C8環烧基、CrCs 鹵烷基、及CrC8烷氧基之取代基所取代; 每一Rs、及116係各自獨立為氫、或G-Cs烷基;Wherein each X and γ system is independently oxygen, Nr5, or sulfur; R is hydrogen, or ¢: (=0) (:, - (: 8 alkyl; each R丨, R2, and R3 Each is independently hydrogen, methyl, or (CH2)m_CH3; R4 is NR5R6, 〇R5, 〇c(=〇)r7, c(=〇)OR5, cdi)r5, halogen, 5 or 6 membered ring Lactone, C|-C8 alkyl, c2-C8 dilute, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, Ci_c8 alkyl, c2_C8 alkenyl, alkynyl, The aryl group and the glucose group are selectively selected from one or more selected from the group consisting of NR5R6, 〇R5, 〇C(=〇)R7, c(=0)OR5, c(=o)r5, C"c8, and c2 Substituted by -c8 dilute, c2-c8 block, C3-C8 cycloalkyl, CrCs haloalkyl, and CrC8 alkoxy; each Rs, and 116 are independently hydrogen, or G-Cs alkyl;

RtSQ-Cs烷棊、〇R5、或NR5R6 ; m為卜12 ;以及 η為 1-12 。 於部分實施例中,主體係為人類。 經研究發現,當誘發疾病7日後,FSGS小鼠展現明顯 的GPx活性提升(如實施例4及14所示)。氧化壓力與其持續 伴隨之發炎反應,係為慢性腎病之普遍特徵(Kim,etal.,j/w J Physiol Renal Physiol, 298: F662-671, 2010; Yoon, et al., /«i, 71: 167-172,2007),且在腎絲球硬化症發展中 扮演重要的角色。需特別注意的是’氧化壓力及發炎反應 30 201247612 彼此之間息息相關,其中任何一者均會誘發以及放大另一 者,而造成惡性循環。舉例而言,氧化壓力可包括活化 NF-κΒ、及伴隨產生之促發炎因子及趨化素(Chemokine)所 造成之發炎反應,因而導致白血球活化及ROS/NO的產生與 釋放,進而造成腎臟中的氧化壓力(Anrather,et al.,价〇/ Chem, 281 : 5657-5667, 2006; Vaziri, et al., Nat Clin Pract Nephrol, 2: 582-593, 2006; Rodrigo, et al., Free Radio Biol Me式33: 409-422, 2002)。此外,相較於正常小鼠,NF-kB 活化及其後續所引發之第二型環氧酵素、誘發型一氧化氮 合成酶、】L-6、及TNF-α,均會造成Nrf2基因缺陷小鼠嚴重 的發炎反應(Chen,et al.,J/w «/ //eari CZrc 290: HI862-1870, 2006; Li, et al., Biochem Pharmacol, 76: 1485-1489,2008)。此外,亦有研究顯示,HO-1缺乏亦會造 成腎絲球硬化,其中HO-1係由Nrf2所調節(Datta,et al.,J dw Soc 从?/7/2广〇/, 10: 2540-2550, 1999)。 於部分實施例中,當投予本發明所提供之環己烯酮化 合物(如:化合物1),可明顯減少腎IL-6表現,並抑制腎臟 中NF-κΒ活化。經FSGS+Antroq小鼠實驗結果,已證實T細 胞及巨噬細胞浸潤至腎臟,可明顯受到抑制(請參見實施例 5、11及14)。於部分實施例中,此效果之作用機制,與抑 制間質性發炎及EPHL相關,其中EPHL為造成腎臟中FSGS 發展之主要因素之一(D'Agati,V Sewi/i 23: 1 17-134, 2003; Nagata, et al., Lab Invest, 80: 869-880, 2000)〇 y y201247612 本發明之部分實施例係提供一種增加腎臟中榖胱甘肽 過氧化酶(GPx)活性之方法,包括:投予一有效劑量之一環 己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、其溶 劑化物 '或其前趨藥物至該主體,其中,該環己烯酮化合 物係具有下列結構:RtSQ-Cs alkane, 〇R5, or NR5R6; m is 卜12; and η is 1-12. In some embodiments, the primary system is a human. It has been found that FSGS mice exhibit a significant increase in GPx activity after 7 days of disease induction (as shown in Examples 4 and 14). Oxidative stress and its associated inflammatory response are common features of chronic kidney disease (Kim, et al., j/w J Physiol Renal Physiol, 298: F662-671, 2010; Yoon, et al., /«i, 71: 167-172, 2007), and plays an important role in the development of renal sclerosing. It is important to note that 'oxidative stress and inflammatory response 30 201247612 are closely related to each other, and either one induces and amplifies the other, causing a vicious circle. For example, oxidative stress can include activation of NF-κΒ, and the associated inflammatory factors and chemotactic reactions caused by Chemokine, resulting in leukocyte activation and ROS/NO production and release, which in the kidneys Oxidative pressure (Anrather, et al., Price / Chem, 281: 5657-5667, 2006; Vaziri, et al., Nat Clin Pract Nephrol, 2: 582-593, 2006; Rodrigo, et al., Free Radio Biol Me 33: 409-422, 2002). In addition, compared with normal mice, NF-kB activation and its subsequent initiation of type 2 epoxidase, induced nitric oxide synthase, L-6, and TNF-α cause Nrf2 gene defects. Severe inflammatory response in mice (Chen, et al., J/w «/ //eari CZrc 290: HI862-1870, 2006; Li, et al., Biochem Pharmacol, 76: 1485-1489, 2008). In addition, studies have shown that HO-1 deficiency can also cause glomerular sclerosis, in which HO-1 is regulated by Nrf2 (Datta, et al., J dw Soc from ?/7/2 〇 /, 10: 2540-2550, 1999). In some embodiments, administration of a cyclohexenone compound (e.g., Compound 1) provided by the present invention significantly reduces renal IL-6 expression and inhibits NF-κΒ activation in the kidney. As a result of FSGS+Antroq mouse experiments, it has been confirmed that T cells and macrophages infiltrate into the kidney and can be significantly inhibited (see Examples 5, 11 and 14). In some embodiments, the mechanism of action is related to the inhibition of interstitial inflammation and EPHL, among which EPHL is one of the major factors contributing to the development of FSGS in the kidney (D'Agati, V Sewi/i 23: 1 17-134). , 2003; Nagata, et al., Lab Invest, 80: 869-880, 2000) 〇y y201247612 Some embodiments of the invention provide a method of increasing glutathione peroxidase (GPx) activity in the kidney, including : administering an effective amount of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中’每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或C(=0)C「C8烧基; 每一 R,、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 NR5R6、〇r5、〇c(=〇)r7、c(=0)0R5、C(=0)R5、鹵 素、5或6員環内酯、CrCe烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Ci-Cs院基、C2_C8 稀基、C2-C8块基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇r5、〇c(=〇)r7、c(=0)〇R5、c(=o)r5、 CVCs烷基、C2-C8烯基、c2-C8炔基、c3-c8環烷基、c!-c8 卤烧基、及Ci-Cs院氧基之取代基所取代; 每一 Rs、及R0係各自獨立為氫 '或C丨·c8烷基; R7係Ci-Cg烧基、or5、或nr5r6 ; m為1 -12 ;以及 η為 1-12 。 於部分實施例中’主體係為人類。 32 201247612 本發明之部分實施例係提供一種減少一主體之促炎細 胞因子之方法’包括:投予—有效劑量之一環己烯酮化合 ’、醫藥上可接受之鹽類、其代謝物、其溶劑化物 '或 其前趨藥物至該主n,其中,該環己稀嗣化合物係具有下 列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or C(=0)C"C8 alkyl; each R, R2, and R3 are each independently hydrogen, Methyl or (CH2)m-CH3; R4 is NR5R6, 〇r5, 〇c(=〇)r7, c(=0)0R5, C(=0)R5, halogen, 5 or 6 membered ring lactone , CrCe alkyl, C2-C8 alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, Ci-Cs, C2_C8, C2-C8, The aryl group and the glucosyl group are selected from one or more selected from the group consisting of NR5R6, 〇r5, 〇c(=〇)r7, c(=0)〇R5, c(=o)r5, CVCs alkyl, C2- Substituted by a C8 alkenyl group, a c2-C8 alkynyl group, a c3-c8 cycloalkyl group, a c!-c8 haloalkyl group, and a substituent of a Ci-Cs alkoxy group; each Rs and R0 are each independently hydrogen' Or C丨·c8 alkyl; R7 is a Ci-Cg alkyl group, or5, or nr5r6; m is 1 -12; and η is 1-12. In some embodiments, the 'main system is human. 32 201247612 Some embodiments provide a method for reducing a pro-inflammatory cytokine of a subject 'including: administration-effective dose of one cyclohexenone compound', pharmaceutically acceptable The salts, their metabolites, solvates' or its predecessors to the main medicament n, wherein the Si compound is cyclohexene column having the structure:

其中,每一X及γ係各自獨立為氧、NR5、或硫; R係為氫、或CPOKVCs燒基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3; R4係為 NR5R6、〇R5、〇C(=〇)R7、C(=〇)〇R5、c(=〇)r5、鹵 素、5或6員環内醋、C!-C8院基、c2-C8稀基、C2-C8块基、 芳基、或葡萄糖基,其中,5或ό員環内酯、烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、〇C(=〇)R7、C(=0)0R5、C(=0)R5、 CVCs院基、C2-C8稀基、c2-c8快基、c3-c8環烧基、C〗-C8 鹵烷基、及CrCs烷氧基之取代基所取代; 每一R5、及R6係各自獨立為氫、或Crc8烷基; RACi-Ce烷基、〇R5、或NR5R6 ; m為1-12 ::以及 η 為 1-12 » 於部分實施例中’主體係為人類。 33 201247612 本發明之部分實施例係提供一種減少腎臟中硫胱胺酸 蛋白酶-1表現及/或抑制腎NLRP3活性之方法,包括:投予 一有效劑量之一環己烯酮化合物、其醫藥上可接受之鹽 類、其代謝物、其溶劑化物、或其前趨藥物至該主體,其 中,該環己烯酮化合物係具有下列結構:Wherein each of the X and γ systems is independently oxygen, NR5, or sulfur; R is hydrogen or CPOKVCs; each R丨, R2, and R3 are independently hydrogen, methyl, or (CH2) m_CH3; R4 is NR5R6, 〇R5, 〇C(=〇)R7, C(=〇)〇R5, c(=〇)r5, halogen, 5 or 6 member ring vinegar, C!-C8 yard, a c2-C8 dilute group, a C2-C8 block group, an aryl group, or a glucosyl group, wherein 5 or a member of the ring lactone, an alkyl group, a C2-C8 alkenyl group, a CrC8 alkynyl group, an aryl group, and a glucosyl group are selected One or more selected from the group consisting of NR5R6, OR5, 〇C(=〇)R7, C(=0)0R5, C(=0)R5, CVCs, C2-C8 dilute, c2-c8 fast radical, Substituted by a substituent of a c3-c8 cycloalkyl group, a C-C8 haloalkyl group, and a CrCs alkoxy group; each R5 and R6 are each independently hydrogen or a Crc8 alkyl group; RACi-Ce alkyl, fluorene R5, or NR5R6; m is 1-12::: and η is 1-12 » In some embodiments, the 'main system is human. 33 201247612 A portion of the present invention provides a method of reducing thiocysteine-1 expression in the kidney and/or inhibiting renal NLRP3 activity, comprising: administering an effective dose of one of the cyclohexenone compounds, which is pharmaceutically acceptable Accepting a salt, a metabolite thereof, a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中,每一 X及Y係各自獨立為氧、NR5、或琉; R係為氫、或烷基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m-CH3 ; R4係為 nr5r6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内酯' Ci-Cs烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、C^-Cs烷基、C2-C8 烯基、C^Cg炔基 '芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、 c】-c8烷基、C2-C8烯基、C2-C8炔基、C3-C8環烷基、CVCs 鹵烷基、及CrCs烷氧基之取代基所取代; 每一r5、及尺6係各自獨立為氮、或c,-c8烷基; ISCVCe烷基、OR5、或NR5R6 ; m為1-12 ;以及 η為 1-12 。 於部分實施例中,主體係為人類。 34 201247612 活性之方法,包括:投予一 其醫藥上可接受之鹽類、; 趨藥物至該主體,其中,言 構: 本發月之刀實施例係提供一種減少腎臟中腎 -有效劑量之一環己烯酮化合物、 其代物、其溶劑化物、或其前 該環己稀嗣化合物係具有下列結Wherein each of the X and Y systems is independently oxygen, NR5, or hydrazine; R is hydrogen or an alkyl group; each of R, R2, and R3 is independently hydrogen, methyl, or (CH2)m -CH3 ; R4 is nr5r6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone 'Ci-Cs alkyl, C2-C8 Alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, C^-Cs alkyl, C2-C8 alkenyl, C^Cg alkynyl 'aryl, and glucose One or more selected from the group consisting of NR5R6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, c]-c8 alkyl, C2-C8 alkenyl, C2 Substituted by a substituent of -C8 alkynyl, C3-C8 cycloalkyl, CVCs haloalkyl, and CrCs alkoxy; each r5, and 6 is independently nitrogen, or c,-c8 alkyl; ISCVCe Alkyl, OR5, or NR5R6; m is 1-12; and η is 1-12. In some embodiments, the primary system is a human. 34 201247612 A method of activity comprising: administering a pharmaceutically acceptable salt thereof; and stimulating a drug to the subject, wherein: the present invention provides a method for reducing renal-effective dose in the kidney a cyclohexenone compound, a substitute thereof, a solvate thereof, or a pre-cyclohexanthene compound thereof having the following

其中’每一 X及Υ係各自獨立為氧、Nr5、或硫; R係為氫'*或CpCOCnCe烷基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或 R4係為 NR5R6、OR5、〇C(=0)R7、c(=〇)〇R5 ' c(=0)R5、鹵 素、5或6員環内醋、Ci-C8^基、c2-C8烯基、c2-c8炔基、 芳基、或葡萄糖基’其中,5或6員環内酯、Cl-C8烷基、c2_C8 烯基、Cr-Cs炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R>6、0R5、〇C(=〇)R7、c(=〇)〇R5、C(=0)R5、 CVCs烧基、C2-C8稀基、c2-c8炔基' c3-C8環院基' CKC8 鹵烷基、及c丨-C8烷氧基之取代基所取代; 每一 R5、及Re係各自獨立為氫、或C|-C8烧基;Wherein 'each X and oxime are each independently oxygen, Nr5, or sulfur; R is hydrogen '* or CpCOCnCe alkyl; each R丨, R2, and R3 are each independently hydrogen, methyl, or R4 Is NR5R6, OR5, 〇C(=0)R7, c(=〇)〇R5 'c(=0)R5, halogen, 5 or 6 membered vinegar, Ci-C8^, c2-C8 alkenyl, a c2-c8 alkynyl, aryl, or glucosyl group wherein 5 or 6 membered cyclo lactone, Cl-C8 alkyl, c2_C8 alkenyl, Cr-Cs alkynyl, aryl, and glucosyl-selective One or more selected from the group consisting of NR5R>6, 0R5, 〇C(=〇)R7, c(=〇)〇R5, C(=0)R5, CVCs alkyl, C2-C8 dilute, c2-c8 alkynyl' Substituted by a substituent of the c3-C8 ring-based group 'CKC8 haloalkyl, and c丨-C8 alkoxy; each R5, and Re are each independently hydrogen or C|-C8 alkyl;

RjCi-C;烷基、〇R5、或 NR5R6 ; m為1-12 ;以及 η 為 1-12。 於部分實施例中’主體係為人類。 35 201247612 本發明之部分實施例係提供一種抑制腎臟中細胞〉周亡 之方法,包括:投予一有效劑量之一環己烯酮化合物其 醫藥上可接受之鹽類、其代謝物、其溶劑化物、或其前趨 藥物至該主體,其中,該環己烯酮化合物係具有下列結構:RjCi-C; alkyl, hydrazine R5, or NR5R6; m is 1-12; and η is 1-12. In some embodiments the 'main system is human. 35 201247612 A portion of the present invention provides a method for inhibiting cell death in the kidney comprising: administering an effective dose of one of the cyclohexenone compounds, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solvate thereof. Or a pre-drug thereof to the host, wherein the cyclohexenone compound has the following structure:

其中’每一 X及γ係各自獨立為氧、nr5、或硫; R係為氫、或烷基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)r7、c(=〇)〇R5、C(=〇)R5、鹵 素、5或6員環内醋、Cl_c8炫基、C2_C8稀基、C2_C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Ci_C8烷基、C2_C8 烯基、C^-C:8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 nr5r6、〇r5、〇c(=〇)r7、c(=o)〇r5、c(=o)r5、 cvc8烧基、(:2-(:8稀基、c2-c8快基、c3-c8環烧基、(VC8 函院基、及Ci-Cs烷氧基之取代基所取代; 每一R5、及Re係各自獨立為氫、或Ci-C8烷基; 烷基、〇R5、或NR5R6 ; m為1-12 ;以及 η為 1-12 。 於部分實施例中,主體係為人類。 本發明之部分實施例係提供一種保護或預防一主體腎 臟罹患腎絲球硬化症及/或腎間質纖維化及/或腎絲球腎炎 36 201247612 之方法,包括:投予一有效劑量之一環己烯酮化合物、其 醫藥上可接受之鹽類'其代謝物、其溶劑化物、或其前趨 藥物至該主體’以減少腎臟中TGF_P 1蛋白之表現程度及膠 原蛋白I、III及IV蛋白聚集’其中,該環己烯酮化合物係具 有下列結構:Wherein each 'X and γ are each independently oxygen, nr5, or sulfur; R is hydrogen or an alkyl group; each R丨, R2, and R3 are each independently hydrogen, methyl, or (CH2)m_CH3 R4 is NR5R6, 〇R5, 〇c(=〇)r7, c(=〇)〇R5, C(=〇)R5, halogen, 5 or 6 member ring vinegar, Cl_c8 炫, C2_C8 rare group, C2_C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, Ci_C8 alkyl, C2_C8 alkenyl, C^-C:8 alkynyl, aryl, and glucosyl selective Or above selected from nr5r6, 〇r5, 〇c(=〇)r7, c(=o)〇r5, c(=o)r5, cvc8 alkyl, (: 2-(:8 dibasic, c2-c8 fast Substituted by a substituent of a c3-c8 cycloalkyl group, (a VC8-complex group, and a Ci-Cs alkoxy group; each R5, and Re is independently hydrogen or a Ci-C8 alkyl group; 〇R5, or NR5R6; m is 1-12; and η is 1-12. In some embodiments, the primary system is human. Some embodiments of the present invention provide protection or prevention of renal spheroid sclerosis in a subject kidney And/or renal interstitial fibrosis and/or renal glomerulonephritis 36 201247612, including: An effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a prodrug thereof to the subject to reduce the expression of TGF_P 1 protein in the kidney and collagen I, III, and IV protein aggregation ' Among them, the cyclohexenone compound has the following structure:

其中’每一X及Y係各自獨立為氧' NR5、或硫; R係為氫•或C(=0)C|-C8炫基; 每一 Ri、R2、及R_3係各自獨立為氫 '甲基、或(CH2)m_CH3 ; R4係為 NR5R6、OR5、〇C(=〇)R7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内醋、CVCs院基、c2-c8烯基、C2-C8炔基、 芳基、或葡萄糖基’其中’5或6員環内酯、烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、or5、〇c(=o)r7、c(=o)or5、c(=o)r5、Wherein 'each X and Y are each independently oxygen 'NR5, or sulfur; R is hydrogen or C(=0)C|-C8 炫; each Ri, R2, and R_3 are independently hydrogen' Methyl, or (CH2)m_CH3; R4 is NR5R6, OR5, 〇C(=〇)R7, c(=o)or5, c(=o)r5, halogen, 5 or 6 member vinegar, CVCs a c2-c8 alkenyl group, a C2-C8 alkynyl group, an aryl group, or a glucosyl group, wherein '5 or 6 membered ring lactones, alkyl groups, C2-C8 alkenyl groups, CrC8 alkynyl groups, aryl groups, and glucosyl groups Selectively one or more selected from the group consisting of NR5R6, or5, 〇c(=o)r7, c(=o)or5, c(=o)r5,

CkCs烷基、C2-C8烯基、C2-C8炔基、c3-c8環烷基、Ci-Cs 鹵烷基、及Ci-Ce烷氧基之取代基所取代; 每一 R5、及116係各自獨立為氫、或Ci-Cs烷基; 烷基、〇R5、或NR5R6 ; m為1-12 ;以及 η為 M2。 於部分實施例中,主體係為人類。 37 201247612 由活性氧(ROS)及/或一氧化氮(NO)產生增加與抗氧化 能力受損所導致之氧化壓力,可能導致壞疽(necrosis)、細 胞凋亡、發炎、纖維化及其他腎臟疾病(靖參見Kim, et al., dm J_P/j少·π·ο/ 尸/2>^〇/,298: F662-671,2010)。近年來 有關腎絲球切片/硬化機制研究發現,腎臟損傷中之浸潤白 血球細胞與腎臟細胞本身的NAD(P)H氧化酶酵素複合物, 與超氧化物產生有高度相關性(IBID; Jones,et al.,J dm Soc Nephrol, 5: 1483-1491, 1995; Radeke, et al., J Biol Chem, 266: 21025-21029, 1991)。抑制氧化壓力可透過抗發炎及抗 細胞凋亡過程而改善腎臟硬化。此外,細胞核因數-E2相關 因數2 (Nrf2)係為一轉錄因子,在多種細胞及組織中,其會 與能解碼出多種抗氧化及第二期酵素之基因的啟動子區域 中之抗氧化反應因子結合,如榖胱甘肽過氧化酶(GPx)、觸 酶(catalase)、及超氧歧化酵素(superoxide dismutase)等(Itoh, et al” Biochem Biophys Res Commun, 236: 313-322, 1997; Nguyen, et al·,CTzew, 284: 13291-13295, 2009)。細胞 抗氧化反應及抗發炎機制之Nrf2相關調節,係與抗氧化壓 力息息相關;而在小鼠腎小管上皮細胞中,Nrf2訊息傳遞 路徑透過轉化生長因子(TGF)-pi相關之上皮間質轉化 (epithelial-mesenchymal transition)而與腎絲球保護相關 (Shin, et al·,Free 5ι·ο/ Mei/, 48: 1051-1063, 2010),且 同時與鏈腺佐菌素(streptozotocin)誘導之糖尿病腎病相關 (Jiang, et al_,59: 850-860, 2010) 〇 纖維化相關細胞因子的表現,特別是TGF-β 1,係與腎 硬化/纖維化高度相關(Ka, et al., «/ dm Soc TVep/iro/,18: 38 201247612 1777-1788, 2007; Lan, HY. Front Biosci, 13: 4984-4992, 2008; Zhao, et al·,d/w «/TVep/iz-o/, 28: 548-554,2008)。目前 已證實,透過減少TGF-βΙ與胞外基質蛋白表現,而可達到 抑制氧化麼力,進而改善腎絲球硬化(Hahn, et al.,尸ecfiair Nephrol, 13: 195-198, 1999; Kashihara, et al., Curr Med C/zem.; Manning, et al·,dw «/25: 3 11-3 17,2005)。 此外,在小鼠腎小管上皮細胞與鏈脲佐菌素誘導之糖尿病 腎病中,與Nrf2轉錄因子及其第二期酵素有關之抗氧化壓 力訊息傳遞,可保護腎纖維化。 於部分實施例_,當投予本發明所提供之環己烯酮化 合物(如:化合物1),可減少腎臟中TGF-βΙ蛋白表現量,以 及其下游之膠原蛋白I、III及IV蛋白聚集(請參見實施例6及 14);此結果代表在FSGS治療小鼠中’’本發明所提供之環 己烯酮化合物可透過抑制TGF-βΙ相關纖維化路徑’保護腎 臟腎絲球硬化及間質纖維化的產生。 本發明之部分實施例係提供一種治療一主體之局部性 腎絲球硬化症(FSGS)之方法,包括:投予一有效劑量之一 環己烯酮化合物 '其醫藥上可接受之鹽類、其代謝物、其 溶劑化物、或其前趨藥物至該主體,以(i)增加Nrf2活性及 /或(Π)抑制腎臟中NF-κΒ相關發炎反應及TGF-βΙ誘導之纖 維化,其中,該環己稀酮化合物係具有下列結構·Substituted by a substituent of a CkCs alkyl group, a C2-C8 alkenyl group, a C2-C8 alkynyl group, a c3-c8 cycloalkyl group, a Ci-Cs haloalkyl group, and a Ci-Ce alkoxy group; each R5, and 116 series Each is independently hydrogen or a Ci-Cs alkyl group; alkyl, hydrazine R5, or NR5R6; m is 1-12; and η is M2. In some embodiments, the primary system is a human. 37 201247612 Oxidative stress caused by increased production of reactive oxygen species (ROS) and/or nitric oxide (NO) and impaired antioxidant capacity, may lead to necrosis, apoptosis, inflammation, fibrosis and other kidney diseases (See Kim, et al., dm J_P/j less. π·ο/ corpse/2>^〇/, 298: F662-671, 2010). In recent years, studies on the skein section/hardening mechanism of kidney have found that the NAD(P)H oxidase enzyme complex of infiltrating white blood cells in kidney injury and kidney cells itself is highly correlated with superoxide production (IBID; Jones, Et al., J dm Soc Nephrol, 5: 1483-1491, 1995; Radeke, et al., J Biol Chem, 266: 21025-21029, 1991). Inhibition of oxidative stress improves kidney hardening through anti-inflammatory and anti-apoptotic processes. In addition, the nuclear factor-E2 correlation factor 2 (Nrf2) is a transcription factor that, in a variety of cells and tissues, is associated with antioxidant responses in the promoter region of genes that encode multiple antioxidant and second-phase enzymes. Factor binding, such as glutathione peroxidase (GPx), catalase, and superoxide dismutase (Itoh, et al) Biochem Biophys Res Commun, 236: 313-322, 1997; Nguyen, et al., CTzew, 284: 13291-13295, 2009). Nrf2-related regulation of cellular antioxidant responses and anti-inflammatory mechanisms is closely related to antioxidant stress; whereas in mouse renal tubular epithelial cells, Nrf2 signaling Pathways are associated with renal spheroid protection through transforming growth factor (TGF)-pi-related epithelial-mesenchymal transition (Shin, et al., Free 5ι·ο/ Mei/, 48: 1051-1063, 2010), and at the same time associated with streptozotocin-induced diabetic nephropathy (Jiang, et al, 59: 850-860, 2010), the performance of sputum fibrosis-associated cytokines, especially TGF-β 1, High with renal hardening/fibrosis Degree correlation (Ka, et al., «/ dm Soc TVep/iro/, 18: 38 201247612 1777-1788, 2007; Lan, HY. Front Biosci, 13: 4984-4992, 2008; Zhao, et al·, d /w «/TVep/iz-o/, 28: 548-554, 2008). It has been confirmed that by reducing the expression of TGF-βΙ and extracellular matrix proteins, inhibition of oxidative stress can be achieved, thereby improving spheroid atherosclerosis. (Hahn, et al., corpus ecfiair Nephrol, 13: 195-198, 1999; Kashihara, et al., Curr Med C/zem.; Manning, et al., dw «/25: 3 11-3 17,2005 In addition, in mouse renal tubular epithelial cells and streptozotocin-induced diabetic nephropathy, the anti-oxidative stress message associated with Nrf2 transcription factor and its second phase enzyme protects renal fibrosis. For example, when a cyclohexenone compound (such as Compound 1) provided by the present invention is administered, the amount of TGF-βΙ protein in the kidney and the aggregation of collagen I, III and IV downstream thereof can be reduced (see Examples 6 and 14); this result represents that the cyclohexenone compound provided by the present invention is permeable to inhibit TGF-βΙ-related fibrosis in FSGS-treated mice. Diameter 'of protection in kidney glomerular sclerosis and interstitial fibrosis. Some embodiments of the present invention provide a method of treating localized renal sclerosing sclerosis (FSGS) in a subject comprising: administering an effective amount of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite, a solvate thereof, or a pre-drug thereof to the host, (i) increasing Nrf2 activity and/or (Π) inhibiting NF-κΒ-related inflammatory response in the kidney and TGF-βΙ-induced fibrosis, wherein The cyclohexanone compound has the following structure.

201247612 其中,每一 χ及Y係各自獨立為氧、NR5、或硫; R係為氫、或CpOKrCs烷基; 每一R,、R2、及R3係各自獨立為氨、甲基、或(CH2)m-CH3 ; R4係為 NR5R6、OR5、oc(=o)r7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内酯、CVC8烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、CrC8烷基、C2-Cs 烯基、C2-C8炔基、芳基、及葡萄糖基係選擇性的經一種成 以上選自由 NR5R6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、 CrC8烷基、C2-C8烯基、c2-c8炔基、C3-C8環烷基、C,-C8 鹵烷基、及crc8烷氧基之取代基所取代; 每一R5、及116係各自獨立為氫、或C丨-c8烷基; ISCi-Ce烷基、〇R5、或NR5R6 ; m為1-12 ;以及 η 為 1 -12。 於部分實施例中,主體係為人類。 本發明之部分實施例係提供一種治療一主體之腎絲球 腎炎之方法’包括:投予一有效劑量之一環己烯酮化合物、 其醫藥上可接受之鹽類、其代謝物'其溶劑化物、或其前 趨藥物至該主體,以(i)抑制腎NLRP3發炎體活化及/或(ii) 抑制T細胞活性增加,其中,該環己烯酮化合物係具有下列 結構:201247612 wherein each of the ruthenium and Y is independently oxygen, NR5, or sulfur; R is hydrogen, or CpOKrCs alkyl; each R, R2, and R3 are each independently ammonia, methyl, or (CH2) m-CH3; R4 is NR5R6, OR5, oc(=o)r7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, CVC8 alkyl, C2-C8 Alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, CrC8 alkyl, C2-Cs alkenyl, C2-C8 alkynyl, aryl, and glucosyl group are selected One or more selected from NR5R6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, CrC8 alkyl, C2-C8 alkenyl, c2-c8 alkynyl, C3 Substituted by a C8 cycloalkyl, C,-C8 haloalkyl, and crc8 alkoxy substituent; each R5, and 116 are each independently hydrogen or C丨-c8 alkyl; ISCi-Ce alkyl , 〇R5, or NR5R6; m is 1-12; and η is 1 -12. In some embodiments, the primary system is a human. Some embodiments of the present invention provide a method of treating renal glomerulonephritis in a subject comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solvate thereof. Or a pre-drug to the subject, (i) inhibiting activation of renal NLRP3 inflammatory body and/or (ii) inhibiting an increase in T cell activity, wherein the cyclohexenone compound has the following structure:

40 201247612 其中’每一 X及Y係各自獨立為氧、nr5、或硫; R係為氫、.或CpOKVCs烷基; 每一 R,、R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、OR5、〇C(=〇)R7、c(=〇)〇R5、C(=0)R5、_ 素、5或6員環内醋、0「<:8烧基、c2-C8稀基、C2-C8炔基、 芳基、或葡萄糖基’其中,5或6員環内酯、(^-<:8烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、or5、oc(=o)r7、c(=〇)OR5、c(=o)r5、 基、C2-C8稀基、c2-c8炔基、c3-C8環院基、CVCe 鹵烷基、及CrC:8烷氧基之取代基所取代; 每一 R5、及R6係各自獨立為氫、或烷基; R7係 C〗-C8院基、〇Κ·5、或nr5r6 ; m為1-12 ;以及 η為 1-12 。 於本發明中,環己烯酮化合物1可改變Τ細胞活性,且 在AcP-IgAN小鼠中預防腎發炎反應,故本發明之示例環己 烯酮化合物適用於減緩IgAN » 本發明之特定實施例’係提供一種減緩一主體之免疫 球蛋白A型腎炎(IgAN)之方法,包括:投予一有效劑量之一 環己烯酮化合物、其醫藥上可接受之鹽類、其代謝物、其 溶劑化物、或其前趨藥物至該主體,其中,該環己烯酮化 合物係具有下列結構: 20124761240 201247612 wherein 'each X and Y are each independently oxygen, nr5, or sulfur; R is hydrogen, or CpOKVCs alkyl; each R, R2, and R3 are each independently hydrogen, methyl, or (CH2)m_CH3 ; R4 is NR5R6, OR5, 〇C(=〇)R7, c(=〇)〇R5, C(=0)R5, _素, 5 or 6 members of the ring vinegar, 0 "< :8 alkyl, c2-C8 dilute, C2-C8 alkynyl, aryl, or glucosyl' wherein 5 or 6 membered cyclic lactones, (^-<:8 alkyl, C2-C8 alkenyl, The CrC8 alkynyl, aryl, and glucosyl group are selected from one or more selected from the group consisting of NR5R6, or5, oc(=o)r7, c(=〇)OR5, c(=o)r5, yl, C2-C8 Substituted by a substituent of a dilute group, a c2-c8 alkynyl group, a c3-C8 ring-based group, a CVCe haloalkyl group, and a CrC:8 alkoxy group; each of R5 and R6 is independently hydrogen or an alkyl group; R7 is C--C8, 〇Κ·5, or nr5r6; m is 1-12; and η is 1-12. In the present invention, cyclohexenone compound 1 can change sputum cell activity, and in AcP -IgAN mice prevent renal inflammatory response, so an exemplary cyclohexenone compound of the invention is suitable for slowing IgAN » specific embodiments of the invention ' A method for alleviating a subject immunoglobulin type A nephritis (IgAN) comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a precursor drug to the host, wherein the cyclohexenone compound has the following structure: 201247612

其中’每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或c(=o)crc8烷基; 每一 R丨' R2、及R3係各自獨立為氫、甲基、或(CH2)m_CH3 ; R4係為 NR5R6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、鹵 素、5或6員環内酯、C,-C8烷基、c2-c8烯基、c2-c8炔基、 芳基、或葡萄糖基,其令’5或6員環内酯、Ci-Cs烷基、c2-c8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇C(=〇)R7、C(=〇)〇R5、C(=〇)R5、 c,-c8燒基、C2-C8稀基、c2-c8块基、〇3-(:8環院基、CrC8 鹵烷基、及Ci-C8烷氧基之取代基所取代; 每一 R5、及Re係各自獨立為氫、或C「C8烷基; R7係 cvc8院基、〇r5、或 nr5r6 ; m為1-12 ;以及 η為 1-12 » 特定專門用語 除非特別註明,本發明所使用之詞棄,包括說明書及 申請專利範圍中所使用之詞彙,均如下所定義。需了解的 是,除非内容清楚明確指出外,於本發明之說明書及申請 圍二:數表示之「一」⑨「該」係同時包含複數 的意思。除了特殊指出外,任何習知方法,包括質譜儀、 歷'HPLC、蛋白質化學技術、生物化學技術、重組腦 42 201247612 技術、及藥理技術均可用於本發明中。再者,除非特別說 明,本發明所使用之「或」或「以及」係意指「及/或」。 再者,「包括」、「包含」及「含有」係意指非限定。此 外,本發明之各段落標題僅用以編排上之目的並非用以 限制本發明之標的》 「烷基」一岡係指脂肪族碳氫基團。烷基可為飽和烷 基基團(意指不含有任一碳-碳雙鍵或碳_碳三鍵),或烷基可 為不飽和烷基基團(意指含有至少一碳·碳雙鍵或碳碳三 鍵)。無論是飽和或不飽和之烷基基團,均可為支鏈或直鏈 之烧基基團。 「烷基」基團可具有1至10個碳原子;即便本發明亦包 含未定義數字範圍之「烷基」,但無論如何,如「丨至⑺」 之數字範圍係表示此範圍中之每一整數;例如,「丨至⑺個 碳原子」係指烷基可由1個碳原子、2個碳原子、3個碳原子 等,且最多由10個碳原子所組成。本發明之化合物之烷基 可以「0丨-(:6烷基」或其他相似定義方式定義。例如(僅用以 說明)’「CrCei烷基」係指烷基鏈上係有一、二、三、四、 五、或六個碳原子。此外,烷基之一實施態樣係選自由甲 基、乙基、丙基、異丙基、正丁基、異丁基、仲丁基 (sec-buty丨)、及丁基(t_butyl)所組成之群組。一般的烷基基 團包括,但不限於:甲基、乙基、丙基、異丙基'丁基、 異丁基、仲丁基、叔丁基、戊基'新戊基、己基、烯丙基、 2-丁烯基、3_丁烯基、環丙基甲基、環丁基甲基、環戊基曱 43 201247612 基、%己基甲基、及其相似基團。於一實施態樣中,完基 係為cvc6烷基。 亞炫•基」sSJ係指一價炫基自由基(divalent alkyl radical)。上述之任一單價烷基基團均可藉由從烷基移除一 第二氫原子而形成一烯基基團。於一實施態樣中,亞烷基 係為c,-c:6亞烷基。於另一實施態樣中,亞烷基係為 亞炫基。一般的亞燒基基團包括,但不限於:_CH2_、 -CH(CH3)- ' -C(CH3)2- ' -CH2CH2- ' -CH2CH(CH3)- ' •CH2C(CH3)2-、-CH2CH2CH2-、-CH2CH2CH2CH2-、及其相 似基團。 於本發明中,「芳基」一詞係指一芳香環,其中形成 芳香環之各原子係為碳原子。芳香環可由五、六、七、八、 九、或超過九個碳原子所形成。此外,芳香環可選擇性的 被取代。於一實施態樣中,芳基係為苯基或萘基。於一實 施態樣中,芳基係為苯基<»於一實施態樣中,芳基係為C6_C⑺ 芳基》依據結構上不同,芳基基團可為一單自由基或一雙 自由基(即,亞芳基)《»此外,於一實施態樣中,亞芳基係為 Ce-Cio亞芳基《亞芳基之例子包括,但不限於·丨2亞笨基 (phenyM,2-ene)、1,3-亞苯基(phenyi_i,3_ene)、及 ι,4-亞笨 基(phenyl- l,4-ene) 〇 「芳香環」一詞係指一具有非定域71電子系統之平面 環,其中非定域子系統係包括牝以個兀電子,且n係為— 整數。芳香環可由五、六、七、八、九、十、或超過十個 原子所組成。此外,芳香環可選擇性的被取代。「芳香環」 44 201247612 一詞包括:碳環芳基(「芳基」,如 「雜芳基」或「雜芳族基團」)基團(二=環芳基(或 「芳香環」一詞亦^ ^ 比啶基)。此外’ —k:)(即,料㈣碳原子對之環)基團f(fUSed-rmg 、…「南」、「齒素」、或「齒化物」-詞係指氟、氣、 溴、或碘。 <日氣虱 「環内醋」一詞係指環酷類,其為同—分 與叛基-COOH之縮合產物。環内_之特徵在於其為一由兩 :或以上碳原子與一個氧原子所組成之閉環且在相鄰於 乳之碳原子上具有一網基團=〇。 雜環」-詞係指環中具有一至四個雜原+之雜芳香 環(即,雜芳基)及雜環烷基團,其中環中之每一雜原子係選 自由0、S及N,且環系中之每一雜環基團係具有4至1〇個原 子,但任一環並不含有兩相鄰之〇或3原子。非芳香族雜環 基團(即’雜環烷基團)之環系中包括僅具有3個原子之基 團,但芳香族雜環基團之環系中需具有至少5個原子。雜環 基團包括苯并稠和環系(benzo fused ring system)。3員雜環 基團之例子為氮丙咬基(aziridinyl)。4員雜環基團之例子為 氮雜環丁烷基(azetidinyl)。5員雜環基團之例子為噻唑基 (thiazoly!)。6員雜環基團之例子為pyridyl基(pyridyl),而1 〇 員雜環基圏之例子為喧琳基(quinolinyl)。非芳香族雜環基 團例子包括:°比洛炫基(pyrrolidinyl)、四氫吱味基 (tetrahydrofuranyl)、二氫°夫喃基(dihydrofuranyl)、四氫售 吩乙基(【€1^11丫(1|"〇1;11丨611丫1)、°惡*坐炫1酮基(〇乂32〇11<1111〇11)^)、四 201247612 i °比喊基(tetrahydropyranyl)、二氫 °比味基 (dihydropyranyl)、四氫1^味基(tetrahydrothiopyranyl)、派咬 基(piperidinyl)、嗎琳基(morpholinyl) ' 硫代嗎琳基 (thiomorpholinyl)、嘆嗔炫基(thioxanyl)、略唤基 (piperazinyl)、乱丙咬基(aziridinyl)、氣雜環 丁炫基 (azetidinyl)、乙氧甲基酸基(oxetanyl)、硫化三亞甲基 (thietanyl)、高略咬基(homopiperidinyl)、°惡庚因基 (oxepanyl)、硫雜丁環基(thiepanyl)、氧雜0丫庚因基 (oxazepinyl)、二 *»丫 庚因基(diazepinyl)、硫雜0丫 庚因基 (thiazepinyl) 、 1,2,3,6- 四 氩 β比咬基 (l,2,3,6-tetrahydropyridinyl)、2-0比洛琳基(pyrrolin-2-yl)、 3-吼洛琳基(pyrrolin-3-yl)、二氫 β引嗓基(indolinyl)、2Η-β比 味基(2H-pyranyl)、4Η-β比味基(4H-pyranyl)、二。惡烧基 (dioxanyl)、1,3-二氧戊環基(1,3-dioxolanyl)、°比"坐琳基 (pyrazolinyl)、二嘆烧基(dithianyl)、二硫戊環基 (dithiolanyl)、二氫。比喃基(dihydropyranyl)、二氫嘆吩基 (dihydrothienyl)、二氫β夫喊基(dihydrofuranyl)、°比"坐烧基 (pyrazolidinyl)、°米。坐淋基(imidazolinyl)、°米"坐炫基 (imidazolidinyl) 、 3-氮雜雙環[3.1.0]己烷基 (3-azabicyclo[3.1.0]hexanyl)、3-氮雜雙環[4.1.0]庚烷基 (3-azabicyclo[4.1.0]heptanyl)、3H-0弓I0朵基(3H-indolyl)、以 及啥喚基(quinoliziny 1)。芳香族雜環基困例子包括:β比咬基 (pyridinyl)、味。全基(imidazolyl)、嘴咬基(pyrimidinyl)、0比 0坐基(pyrazolyl)、。塞。圭基(triazolyl)、°比嗪基(pyrazinyl)、四 46 201247612 唑基(tetrazolyl)、呋喃基(furyi)、噻吩基(thienyi)、異噁唑 基(isoxazolyl)' 噻唑基(thiazolyl)、唑基(oxazolyl)、異噻唑 基(isothiazolyl)、吡咯基(pyrrolyl)、喹啉基(quinolinyl)、異 喹啉基(isoquinolinyl)、吲哚基(indolyl)、苯并咪唑基 (benzimidazolyl)、苯并呋喃基(benzofuranyl)、噌嗪基 (cinnolinyl)、吲唑基(indazolyl)、吲哚嗪(indolizinyl)、酞嗪 基(phthalazinyl)、哒。秦基(pyridazinyl)、三嗪基(triazinyl)、 異氛茚基(isoindolyl)、蝶咬基(pteridinyl)、嗓吟基 (purinyl)' 噁二唑基(oxadiazolyl)' 噻二唑基(thiadiazolyl)、 0夫0丫基(furazanyl)、苯并咬。丫基(benzofurazanyl)、苯并售吩 基(benzothiophenyi)、苯并售唾基(benzothiazolyl)、苯并嗔 °坐基(benzoxazolyl)、啥。坐淋基(quinazolinyl)、喧喔琳基 (quinoxalinyl)、萘咬基(naphthyridinyl)、及吱喊并"比咬基 (furopyriclinyl)。如有可能,前述基團可能以C-連接或以N-連接。例如,衍生於吡咯之基團可為吡咯-1-基(N-連接)、 或吡咯-3 .基(C-連接)》再者,衍生於咪唑之基團可為咪唑-1-基或咪唑-3-基(皆為N-連接),或咪嗤-2-基、味吐-4-基或咪 唑·5-基(皆為C-連接)。雜環基團係包括苯并稠和環系。# 芳香族雜環則可經一個或兩個氧基團(=0)所取代,如吡咯 琳-2-銅(p:yrrolidin"2-one) 0 合併療法 一般而言,本發明所述之組成物(合併治療所使用之組 成物)、及其他藥劑,並不一定需要以同一醫藥組成物進行 投藥,且於部分實施例中,因藥劑之物理與化學特性步同, 47 r 201247612 而以不同路徑投藥。於部分實施射,第—次投藥係根據 已知的心藥方式,而後臨床醫師將會依據所觀察到的效 果’以修改投藥的劑量與形式及投藥次數。 於部分實施例中,當藥物進行合併治療時,可改變有 效治療劑量。此外,合併治療更包括起始與結束間多次給 藥之間隔治療,而有助於患者臨床治療管理。於本發明中 所述之合併療法,共投予化合物劑量可依照所使用之複合 藥物形式、所使用之特定藥物、疾病、不適情況、或所治 療之病症而加以改變。 應了解的是,於部分實施例中,為了達到舒緩而用以 治療、預防或改善症狀之藥物治療,可依照各種不同因素 而改變。這些因素包括··患者主體所感染的疾病,以及患 者主體之年齡、體重、性別、飲食'及用藥情形。因此, 於其他實施例中,藥物治療實際變化很大,也因此本發明 所述之藥物治療方式也會有所改變。 混合有化合物(即,本發明所述之環己烯酮化合物)及 其他固酵類及/或免疫抑制劑亦包含在本發明中。於部分實 施例令,免疫抑制劑包括’但不限於:醣皮質固醇 (Glucocorticoids)、細胞穩定劑(cytostatics)、抗趙、作用於 免疫素(immunophilin)之藥物、及其他藥物,如干擾素 (interferon)、鸦片(interferon)、TNF結合蛋白及霉酚酸酯 (mycophenolate) 〇 本發明部分實施例係提供一種早期治療腎病(包括腎 絲球硬化症、腎絲球腎炎、及其相似疾病)之醫藥組成物, 48 201247612 包括:一有效劑量之一環己烯酮化合物、其醫藥上可接受 之鹽類、其代謝物、其溶劑化物、或其前趨藥物;以及一 種或以上固醇類及/或免疫抑制劑,其中,該環己烯鲷化合 物係具有下列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or c(=o)crc8 alkyl; each R丨' R2, and R3 are each independently hydrogen, methyl Or (CH2)m_CH3; R4 is NR5R6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, C,-C8 alkane a c2-c8 alkenyl group, a c2-c8 alkynyl group, an aryl group, or a glucosyl group, which is a '5 or 6 membered ring lactone, a Ci-Cs alkyl group, a c2-c8 alkenyl group, a CrC8 alkynyl group, an aryl group. And one or more selected from the group consisting of NR5R6, 〇R5, 〇C(=〇)R7, C(=〇)〇R5, C(=〇)R5, c,-c8 alkyl, C2 Substituting -C8 dilute, c2-c8 block, 〇3-(: 8-ring, CrC8 haloalkyl, and Ci-C8 alkoxy; each R5, and Re are each independently hydrogen Or C "C8 alkyl; R7 is cvc8, 〇r5, or nr5r6; m is 1-12; and η is 1-12 » specific terminology, unless otherwise stated, the term used in the present invention, including instructions And the terms used in the scope of the patent application are defined as follows. It should be understood that the present invention is The book and the application for the second: the number "1" 9 "this" also includes the plural meaning. Unless otherwise specified, any conventional methods, including mass spectrometry, calendar 'HPLC, protein chemistry, biochemical technology, Recombinant Brain 42 201247612 Technical and pharmacological techniques can be used in the present invention. Further, unless otherwise stated, the use of "or" or "and" means "and/or". And "including" are meant to be non-limiting. In addition, the headings of the various paragraphs of the present invention are for illustrative purposes only and are not intended to limit the subject matter of the present invention. The alkyl group may be a saturated alkyl group (meaning that it does not contain any carbon-carbon double bond or carbon-carbon triple bond), or the alkyl group may be an unsaturated alkyl group (meaning that it contains at least one carbon) · Carbon double bond or carbon-carbon triple bond). Whether it is a saturated or unsaturated alkyl group, it may be a branched or linear alkyl group. The "alkyl" group may have 1 to 10 carbons. Atom; even if the invention also includes an "alkyl" group of undefined numerical ranges In any case, the numerical range such as "丨 to (7)" means each integer in the range; for example, "丨 to (7) carbon atoms" means that the alkyl group may be 1 carbon atom, 2 carbon atoms, 3 A carbon atom or the like, and composed of at most 10 carbon atoms. The alkyl group of the compound of the present invention may be defined by "0丨-(:6 alkyl group) or other similar definitions. For example (for illustration only) '"CrCei alkane By "alkyl" is meant one, two, three, four, five, or six carbon atoms in the alkyl chain. Further, one embodiment of the alkyl group is selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-buty(R), and butyl (t-butyl). The group that makes up. Typical alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl 'butyl, isobutyl, sec-butyl, tert-butyl, pentyl' neopentyl, hexyl, alkene Propyl, 2-butenyl, 3-butenyl, cyclopropylmethyl, cyclobutylmethyl, cyclopentyl hydrazine 43 201247612 base, % hexylmethyl, and the like. In one embodiment, the complete system is a cvc6 alkyl group. "Asian base" sSJ refers to a divalent alkyl radical. Any of the above monovalent alkyl groups can form an alkenyl group by removing a second hydrogen atom from the alkyl group. In one embodiment, the alkylene group is c, -c: 6 alkylene. In another embodiment, the alkylene group is a stannylene group. Typical mercapto groups include, but are not limited to: _CH2_, -CH(CH3)- '-C(CH3)2- '-CH2CH2- '-CH2CH(CH3)- ' •CH2C(CH3)2-,- CH2CH2CH2-, -CH2CH2CH2CH2-, and the like. In the present invention, the term "aryl" means an aromatic ring in which each atomic system forming an aromatic ring is a carbon atom. The aromatic ring may be formed by five, six, seven, eight, nine, or more than nine carbon atoms. In addition, the aromatic ring can be optionally substituted. In one embodiment, the aryl group is phenyl or naphthyl. In one embodiment, the aryl group is a phenyl group. In one embodiment, the aryl group is a C6_C(7) aryl group. The aryl group may be a single radical or a pair of free radicals. Base (ie, arylene) "» In addition, in one embodiment, the arylene system is a Ce-Cio arylene. Examples of arylene include, but are not limited to, 丨2 phenyM, 2-ene), 1,3-phenylene (phenyi_i, 3_ene), and ι,4-phenylene (phenyl- l,4-ene) 〇 "aromatic ring" refers to a non-localized 71 A planar ring of an electronic system in which the non-localized subsystem includes a single electron and the n is an integer. The aromatic ring may consist of five, six, seven, eight, nine, ten, or more than ten atoms. In addition, the aromatic ring can be optionally substituted. "Aromatic ring" 44 201247612 The term includes: carbocyclic aryl ("aryl", such as "heteroaryl" or "heteroaromatic group") group (di-cyclic aryl (or "aromatic ring") The word is also ^ ^ pyridine group). In addition, '-k:) (ie, the ring of the (four) carbon atom pair) f (fUSed-rmg, ... "South", "dentate", or "dentate" - The term “fluorine, gas, bromine, or iodine.” The term “in-loop vinegar” refers to the ring-cooled class, which is a condensation product of homo- and rebel-COOH. It is a closed loop composed of two or more carbon atoms and one oxygen atom and has a network group = 〇 on a carbon atom adjacent to the milk. Heterocyclic"-word system has one to four hybrids in the ring + a heteroaromatic ring (ie, a heteroaryl group) and a heterocycloalkyl group, wherein each hetero atom in the ring is selected from the group consisting of 0, S, and N, and each of the heterocyclic groups in the ring system has 4 to 1 〇 atom, but any ring does not contain two adjacent 〇 or 3 atoms. The ring system of the non-aromatic heterocyclic group (ie, 'heterocycloalkyl group) includes a group having only 3 atoms. Aromatic heterocyclic group The ring system is required to have at least 5 atoms. The heterocyclic group includes a benzo fused ring system. An example of a 3-membered heterocyclic group is an aziridinyl group. An example of a group is azetidinyl. An example of a 5-membered heterocyclic group is thiazoly!. An example of a 6-membered heterocyclic group is pyridyl (pyridyl), and 1 member An example of a heterocyclic hydrazine is quinolinyl. Examples of non-aromatic heterocyclic groups include: pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl ), tetrahydrofuran sold phenethyl ([11&11;(1|"〇1;11丨611丫1), °恶*坐炫1 ketone group (〇乂32〇11<1111〇11)^ ), four 201247612 i ° tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl 'thio? Thiomorpholinyl, thioxanyl, piperazinyl, aziridinyl, azadipine (azetidi) Nyl), oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxa Oxepinpinyl, diazinyl, thiazepinyl, 1,2,3,6-tetra-argon beta ratio bite (l, 2, 3) , 6-tetrahydropyridinyl), 2-0 pyrololin-2-yl, 3-pylorin-3-yl, dihydro-β-indolinyl, 2Η-β 2H-pyranyl, 4Η-β aryl (4H-pyranyl), II. Dioxanyl, 1,3-dioxolanyl, ° ratio "pyrazolinyl, dithianyl, dithiolanyl ), dihydrogen. Dihydropyranyl, dihydrothienyl, dihydrofuranyl, ° ratio "pyrazolidinyl, ° meter. Imidazolinyl, °m" imidazolidinyl, 3-azabicyclo[3.1.0]hexane (3-azabicyclo[3.1.0]hexanyl), 3-azabicyclo[4.1 .0] heptyl (3-azabicyclo [4.1.0] heptanyl), 3H-0 indole (3H-indolyl), and quinoliziny 1 (quinoliziny 1). Examples of aromatic heterocyclic groups include: β-pyridinyl, taste. Iridazolyl, pyrimidinyl, 0 to 0 pyrazolyl. Plug. Triazolyl, pyrazinyl, tetra 46 201247612 tetrazolyl, furyi, thienyi, isoxazolyl' thiazolyl, azole Oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, isoquinolinyl, indolyl, benzimidazolyl, benzo Benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, anthraquinone. Pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl 'oxadiazolyl' thiadiazolyl 0, furazanyl, benzo bite. Benzofurazanyl, benzothiophenyi, benzothiazolyl, benzoxazolyl, hydrazine. Sitting on quinazolinyl, quinoxalinyl, naphthyridinyl, and shouting "furopyriclinyl. If possible, the aforementioned groups may be C-linked or N-linked. For example, the group derived from pyrrole may be pyrrol-1-yl (N-linked), or pyrrole-3-yl (C-linked). Further, the group derived from imidazole may be imidazol-1-yl or Imidazolyl-3-yl (all N-linked), or imidaz-2-yl, tat-4-yl or imidazo-5-yl (all C-linked). Heterocyclic groups include benzo-fused ring systems. # aromatic heterocyclic ring may be substituted by one or two oxygen groups (=0), such as pyrroline-2-copper (p:yrrolidin" 2-one) 0 combined therapy, generally speaking, the invention The composition (the composition used in combination therapy), and other agents do not necessarily need to be administered with the same pharmaceutical composition, and in some embodiments, due to the physical and chemical properties of the agent, 47 r 201247612 Different routes of administration. In the case of partial injection, the first administration is based on the known method of cardiac medicine, and the clinician will then modify the dosage and form of administration and the number of administrations based on the observed effect. In some embodiments, the effective therapeutic dose can be varied when the drug is combined. In addition, the combined treatment includes interval treatment between multiple doses at the beginning and end, which helps the patient to manage clinical treatment. In the combination therapy described in the present invention, the total dose of the compound to be administered may be varied depending on the form of the compound to be used, the particular drug used, the disease, the condition of the disorder, or the condition to be treated. It will be appreciated that in some embodiments, the medical treatment used to treat, prevent or ameliorate symptoms for soothing may vary depending on a variety of factors. These factors include the disease that the subject's subject is infected with, and the age, weight, sex, diet, and medication of the subject. Therefore, in other embodiments, the medical treatment actually varies greatly, and thus the treatment method of the present invention may also be changed. The compound (i.e., the cyclohexenone compound of the present invention) and other solid enzymes and/or immunosuppressive agents are also included in the present invention. In some embodiments, immunosuppressive agents include, but are not limited to, Glucocorticoids, cytostatics, anti-Zhao, drugs that act on immunophilins, and other drugs, such as interferons. (interferon), opport (interferon), TNF-binding protein, and mycophenolate. Some embodiments of the present invention provide an early treatment for nephropathy (including glomerulosclerosis, renal glomerulonephritis, and the like) Pharmaceutical composition, 48 201247612 includes: an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a prodrug thereof; and one or more sterols and / or an immunosuppressive agent, wherein the cyclohexene oxime compound has the following structure:

其中,每一 X及Y係各自獨立為氧、NR5、或硫;R係為氫、 或C(=0)C,-C8烷基;每一R广R2、及R3係各自獨立為氫、 甲基、或(CHdm-CHs ; R4係為 NR5R6、OR5、〇C(=〇)R7、 C(=0)0R5、C(=0)R5、鹵素、5或6員環内醋、Ci-Cg炫基、 C2_Cs稀基' C2_C8块基 '芳基、或葡萄糖基,其中,5或6 員環内酯、C「C8烷基、C2-C8烯基、C2-C8炔基、芳基、及 葡萄糖基係選擇性的經一種或以上選自由NR5R6、〇r5、 oc(=o)r7、c(=o)or5、C(=0)R5、Cl-c8烧基、c2_c8稀基、 C2-C8炔基、CVC:8環烷基、c丨-Cs鹵烷基、及c丨-C8烷氧基之 取代基所取代;每一 R5、及R0係各自獨立為氫、或€1(:8烷 基;R7係(:丨-C8烷基、〇R5、或 NR5R6; m為 1-12;以及n為 1_12。 於部分實施例中,腎病係為腎絲球硬化症(如,FSGS中之腎 纖維化或硬化)。於其他實施例中,腎病係為腎絲球腎炎 (如,免疫球蛋白A型腎絲球腎炎(igAN))。 酿皮質固醇」係指固醇荷爾蒙之一類,其可盘荷爾 蒙糖皮質素受體(glucocorticoid receptor, GR)結合。酶皮質 固醇一詞係源自其在葡萄糖代謝中之調節作用、其係合成 49 201247612 於腎上腺皮質(adrenal cortex)中、且其類固醇結構。聽皮質 固醇之例子包括,但不限於:皮質醇(hydrocortisone, Cortisol)、可的松醋酸醋(cortisone acetate)、潑尼松 (prednisone)、潑尼松龍(prednisolone)、曱基潑尼松龍 (methylprednisolone)、德沙美松(dexamethasone)、倍達邁松 (betamethasone)、敗羥潑尼松龍(triamcinolone)、倍氣米松 (beclometasone)、醋酸氟氫可的松(fludrocortisone acetate)、醋酸去氧皮質明(deoxycorticosterone acetate, DOCA)、及搭固嗣(aldosterone)。 作用於免疫素之藥物例子包括,但不限於:環孢素 (cyclosporin)、他克莫司(tacrolimus)、勒克斯(voclosporin) 及其他約調碟酸酶抑制劑(calcineurin inhibitor)、及雷帕撤 素(sirolimus) β 醫藥组成物/劑型 「可接受」一詞係指本發明之藥劑、組成物、或活性 成分不會對治療主體之日常生活健康造成持續有害反應。 牛樟芝係屬薄孔菌科(Meripilaceae)之真菌類。牛樟芝 子實體一般係呈扁平狀或由生長表面向外生長,且子實層 係朝向外;此外,其邊緣可能會略為翹起,而呈現小括弧 狀。大多數的牛樟芝物種均生長在北部溫帶林中,且造成 樹木之褐腐病(brown rot)。此外,此種真菌中之部分特殊物 種更具有療效,而在台灣常作為中藥藥材之一。 於本發明中,「載體」一詞係指相對無毒之化合物或 化學試劑,其可幫助將一化合物送至細胞或組織中。 50 201247612 於本發明中,「共投予」一詞係包括投予所選擇之治 療藥劑至一患者,且更包括將藥劑以相同或不同投藥方 式、或於相同或不同投藥時間之治療方法。 「稀釋劑」一詞係指在投藥之前用以將所使用之化合 物稀釋之化合物。稀釋劑因可提供一更加穩定的環境故 亦可用以穩定化合物。本技術領域常用之鹽類緩衝溶液(其 可用以控制或維持pH值)亦可作為稀釋劑,其包括但不限於 碌酸鹽緩衝溶液。 於本發明中,「有效劑量」或「治療上有效劑量」一 詞意指一投予試劑或化合物之適當劑量,其可於某種程度 上減輕所治療疾病或症狀之一種或以上病癥。藉此,可減 緩及/或緩和疾病之症候、病癥、或病因,或可依需求對生 理^統造餘何其他的改變L治療上使用之「有效 劑量」係指對疾病病癒可提供臨床上顯著改善所需之組成 物劑量’其中組成物係、包含本發明所揭示之化合物。此外, 於不同個賴況下,適當的「有效」劑量係依照如劑量遞 增試驗之方法來決定。 於本發明中,「增進咨「 增強」一詞係指增加或延 長預定功效之效力或持續時間。^ 貝吋間因此,對治療藥劑之增強 效果而言,「增進」或「擗% ^„ 9強」一词係指無論對效力或持 續時間可達到增加或延長之龍,或者是對—线中之其 他治療藥劑可達到増加或延長之效果。此外,本發明所使 用之「增強有效劑量」係指可左祐—么 T相』在一預定系統中提升另一治 療藥劑效果之適當劑量。 5] 201247612 於本發明中,「代謝物」一詞係指化合物經代謝後所 形成之化合物衍生物β「活性代謝物」一詞係指化合物經 代謝後所形成之生理上具有活性之化合物衍生物。此外, 於本發明中’ 「代謝」一詞係指一特定成分經一有機體改 變之所有過程(其包括,但不限於:水解反應及酵素催化反 應)。因此’酵素可對一化合物做特殊的結構改變。例如, 細胞色素Ρ450可催化多種氧化及還原反應,而尿苷二磷酸 葡萄糖裕駿基轉移酶(uridine diphosphate glucuronyltransferase)則可催化將活性葡萄醛酸(glucuronic acid)分子轉移至芳香醇、脂肪醇、羧酸、胺及巯基 (sulphydryl)自由基困上。於本發明中,化合物之代謝物亦 選擇性的可透過:將化合物投予一宿主並分析該宿主之組 織樣品、或將化合物與肝臟細胞體外培養並分析所得之化 合物等方式來定義。 於本發明中’ 「醫藥組合物(pharmaceutical combination)」一詞意指將一種以上活性成分混合或組合所 形成之產物,且包含活性成分之固定及非固定組合物。其 中,「固定組合物」一詞意指將活性成分(一化合物,如本 發明所述之環己炜酮化合物)及一助劑以單一物體或劑型 同時投予至一患者。「非固定組合物」一詞意指將活性成 分(一化合物,如本發明所述之環己烯酮化合物)及一助劑以 分開劑型同時、一起、或無特定時間間隔限制依序投予至 一患者,其中,這樣的投藥方式可提供兩化合物之有效劑 52 201247612 量至患者體内。此外,非固定組合物亦可用於雞尾酒療法 上,如:投予三種或以上活性成分。 「醫藥組成物(pharmaceutical composition)」一詞係指 一化合物(即本發明所述之環己烯酮化合物)與其他化學成 分之混合物’其他化學成分係如載體、穩定劑、稀釋劑、 分散劑、懸浮劑、增稠劑、及/或賦形劑。醫藥組成物可幫 助化合物投予至一物種。本發明所屬技術領域已知之多種 投予化合物之技術包括,但不限於:靜脈注射、口服、喷 霧投藥、腸道外投藥(非消化道投藥)、經眼部投藥、肺部投 藥(吸入投藥)、及外用投藥等。 此外,「主體」或「患者」一詞係包括哺乳動物;而 哺乳動物之例子包括,但不限於哺乳網之任何物種:人類、 如猩猩及其他猿及猴物種等非人靈長類;農場動物,如牛、 馬、羊、山羊、豬;家畜,如兔子、狗、及猫;包括蓄齒 目動物之實驗用動物’如大鼠、小鼠、天竺鼠、及其他相 似動物。於一實施例中,哺乳動物係為人類。 於本發明中,「治療」一詞係包括:透過預防及/或治 療以緩和、減輕或改善疾病或症狀之至少一病癥預防其 他病癥’抑制疾病或症狀(如,防止疾病或症狀產生),緩和 疾病或症狀’使㈣或症狀復L錢或症狀所導致 之症狀,或抑止疾病或症狀之病癥。 投藥途徑 但不限於:經口服、 眼部、肺部、黏膜、 本發明所適用之投藥途徑包括, 靜脈注射、腸道、喷霧式、腸道外、 201247612 皮膚表面、陰道、耳部、鼻腔、及外用投藥等方式。此外, 腸道外投藥之例子包括:肌肉内、皮下、靜脈内、脊趙内 注射’且亦包括腦脊髓膜内、直接腹腔内、腹膜内淋巴 内、鼻腔内注射。 於特疋實施例中,本發明所述之化合物通常可製備成 儲存劑型或持續釋放劑型’且透過局部方式而非系統性方 式投藥,例如,將化合物直接注射至臟器。於特定實施例 中,可透過殖入方式(如皮下或肌肉内)或肌肉内注射等方 式,以投予長效劑型之藥物。再者,於其他實施例中,藥 物可藉由標乾藥物傳遞系統(例如,包覆有器官專一性抗趙 之微脂體)進行投藥。於此實施例中,微脂體可針對一器官 且被此器官選擇性的吸收。於其他實施例中,本發明所述 之化合物可製備成快速釋玫劑型、緩慢釋放劑型、或立即 釋放劑型。於其他實施例中,本發明所述之化合物可以外 用方式投藥。 醫藥组成物/命[型 於部分實施例中,本發明所述之化合物可製備成一醫 藥組成物。於特定實施例中,醫藥組成物可使用一種或以 上生理上可接受之載體並以習知方式製備;且此生理上可 接受之載體包括賦形劑及辅劑,其可幫助活性化合物之加 工製備製程,以製作出醫藥上可用之藥劑。適當的劑型係 依照投藥路徑做選擇。任何醫藥上可接受之技術、載體、 及賦形劑均可適用於製備本發明所述之醫藥組成物: Remington. The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 54 201247612 1995); Hoover, John E., Remington's Pharmaceutical •Scz.ewce·?,Mack Publishing Co., Easton,Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkinsl999)。 本發明係提供一種醫藥組成物,其包括一化合物(如, 本發明所述之環己烯酮化合物)及一醫藥上可接受之稀釋 劑、賦形劑、或載體《於特定實施例中,所述之化合物可 以混合有-化合物(如,本發明所述之環己烯酮化合物)及其 他活性成分之醫藥組成物,以合併用藥治療方式投藥。本 發明包括所有本發明說明書中有關合併治療法段落中所述 之活性成分之組合物。於特定實施例中,醫藥組成物係包 括一種或以上化合物(如,本發明所述之環己烯酮化合物)。 本發明之醫藥組成物係指一化合物(如,本發明所述之 環己烯酮化合物)與其他化合物之混合物,其他化合物係如 載體、穩定劑、稀釋劑、分散劑、懸浮劑、增稠劑、及/或 賦形劑。於特定實施例中,醫藥組成物可有助於將化合物 投予至一生物體。於部分實施例中,本發明之治療方法, 係將治療上有效劑量之化合物(如,本發明所述之環己烯酮 化合物)以一醫藥組成物方式投藥至一具有疾病或症狀之 治療主體。於特定實施例中,哺乳動物係為人類。於特定 實施例中,治療上有效劑量係依照疾病嚴重程度、主體年 齡及徤康程度、化合物效力及其他因素而改變。此外,本 55 201247612 發明所述之化合物可單獨使用,或是可與一種或以上治療 劑(作為混合物之成分)合併使用。 於一實施例中,化合物(如,本發明所述之環己烯酮化 合物)係製備成水溶液形式。於特定實施例中,水溶液之例 子可選自由水、生理上相容之緩衝液(如漢克氏溶液(Hank,s solution)、林格氏溶液(Ringer’s solution)、或生理食鹽水。 於其他實施例中,化合物(如,本發明所述之環己烯酮化合 物)可製備成黏膜方式投藥之劑型。於特定實施例中,黏膜 投藥劑型包括可適當滲透黏膜屏障之滲透劑。於再其他實 施例中,本發明所述之化合物係製備成其他腸道外注射之 劑型’且適當的劑型包括水性或非水性溶液。於特定實施 例中,這類溶液包括生理上相容之緩衝液及/或賦形劑。 於另一實施例中’本發明所述之化合物可製備成口服 劑型。在此,包含本發明所述之化合物(即,本發明所述之 環已烯酮化合物)之製備,係將活性化合物與如醫藥上可接 又之載趙或賦形劑混合。於不同的實施例中,本發明所述 之化合物可製備成口服劑型,其例子包括錠劑、粉末、藥 片、藥丸、膠囊、藥水、凝膠、含藥糖漿、酏劑、藥衆、 懸浮劑、及相似劑型。 於特定實施例中’ 口服劑型製備可將一種或以上固態 賦形劑與一種或以上本發明所述之化合物混合,並選擇性 的研磨所形成之混合物,若需要可於添加適當輔劑後,再 加工粉末混合物以製得錠劑或藥丸核心。特別是,適當的 賦形劑特別為:填充劑,如包含乳糖、蔗糖、甘露糖醇、 56 201247612 或山梨糖醇之糖類;纖維素製劑’如玉蜀黍澱粉、小麥殿 粉、米澱粉、馬鈴薯澱粉、明膠、橡膠樹膠、曱基纖維素、 微晶纖維素、羥丙基曱基纖維素、羧甲基纖維素鈉;或其 他如聚乙烯吡咯烷鲷(PVP或聚維酮)或磷酸鈣β於特定實施 例中’可選擇性的添加崩解劑。崩解劑的例子可包括:交 聯甲竣纖維素納(Croscarmellose sodium)、聚乙稀〇比洛烧 酮、瓊脂、或海藻酸或其鹽類(如海藻酸鈉)。 於一實施例中’如藥丸核心及錠劑之藥劑可做一層或 以上之適當包覆層。於特定實施例中,濃縮糖類溶液係用 以包覆藥劑。糖類溶液可選擇性的包含··其他添加成分, 例如阿拉伯膠、滑石、聚乙烯吡咯烧酮 '卡伯波凝膠 (carbopol gel)、聚乙二醇、及/或二氧化鈦;包覆溶液;以 及適當有機溶劑或溶劑混合物β染料及/或色素亦可選擇性 的添加至包覆層中,以用於辨識藥劑用。此外,染料及/或 色素亦可選擇性使用,以用於標示不同活性化合物藥劑之 組合物。 於特定實施例中,本發明所述之至少一化合物之治療 上有效劑量係可製備成口服劑型以外的形式。口服劑型包 含由明膠所製成之推入套合式膠囊化⑽^行^叩阳丨幻’以及 由明膠及塑化劑(如甘油或山梨糖醇)所製成之軟且密封之 膠囊。於特定實施例中,推入套合式膠囊包含與一種或以 上填充劑混合之活性成分。填充劑之例子可包括:乳糖、 如澱粉之黏著劑、及/或如滑石或硬脂酸鎂之潤滑劑,並選 擇性的包括穩定劑。於其他實施例令,軟膠囊係包括一種 57 201247612 或以上溶解或懸浮在適當液體中之活性化合物。適當液體 之例子包括:一種或以上脂肪油、液態石臘、或液態聚乙 二醇。此外’可選擇性的添加穩定劑。 於其他實施例中,本發明所述之至少一化合物之治療 上有效劑量係可製備成口頰投藥劑型或舌下投藥劑型的形 式。口頻或舌下投藥之可適用劑型例子,包括錠劑、藥片、 或凝膠。於再其他實施例中,本發明所述之化合物可製備 成腸道外注射劑型’其包括適用於大劑量注射(b〇lus injection)或持續灌注之劑型。於特定實施例中,注射劑型 係為單位劑量形式(如,安瓶形式)、或多劑量包裝。此外, 可選擇性的於注射劑型中添加防腐劑。於再其他實施例 中’化合物(即,本發明所述之環己烯酮化合物)之醫藥組成 物係溶於油性或水性媒介物中,而製備成無菌懸浮液、溶 液、或乳化劑等適合用於腸道外注射之劑型。腸道外注射 劑型可選擇性的包括如懸浮劑、穩定劑、及/或分散劑等配 方劑。於特定實施例中,腸道外投藥之醫藥劑型包括水溶 性形式之活性化合物水溶液。於其他實施例中,活性化合 物之懸浮液係製備成適當的油性注射懸浮液β用於本發明 之醫藥組成物中之適當油溶溶劑或媒介之例子,可包括如 之麻油之脂肪油、或如油酸乙醋(ethyi eleate)或三酸甘油醋 (triglyceride)之合成脂肪酸酯、或微脂體》於特定實施例 中’水性注射懸浮液包含可增加懸浮液黏度之物質,如羧 曱基纖維素鈉、山梨糖醇、或葡聚糖。此外,懸浮液可選 擇性的包括適當的穩定劑或試劑,以提升化合物之溶解 58 201247612 度,而用以製備高濃縮溶液。另一方面,於其他實施例中, 活性成分可製成粉末形式,而可在使用前與適當媒介(如無 菌無熱原水(sterile pyrogen-free water))混合。 於一實施態樣中,化合物(即,本發明所述之環己烯酮 化合物)係以本發明所述之方法或本技術領域已知之方法 製備成腸道外注射之溶液,且使用一自動注射器投藥。自 動注射劑係如美國專利第4,031,893號、第5,358,489號、第 5,540,664 號、第 5,665,〇71 號、第 5,695,472 號、及 W0/2005/087297中所揭示’且上述每一篇專利所揭示之内 容均併入本發明以供參考。一般而言,所有的自動注射器 係包括一待注射髋積之溶液,此溶液係包含一化合物(即, 本發明所述之環己烯酮化合物此外,自動注射器係包 括:一用以容置溶液之儲存空間’此儲存空間係流體連通 至一針頭以進行給藥;以及自動插入針頭機構,其係將針 頭插至一患者上以將藥劑傳送至患者體内。例如,注射器 可k供約0.3 mL、0.6 mL、1.0 mL、或其他適合體積之溶液, 且母1 mlL溶液中含有濃度約為〇 5 mg至50 mg之化合物 (即,本發明所述之環己烯酮化合物)》此外,每一注射器可 僅傳送一劑量之化合物。 於再其他實施例中,化合物(即,本發明所述之環己稀 酮化合物)可外用投藥》在此,本發明所述之化合物可製備 成各種可外用投藥之組成物,如溶液、懸浮液、乳劑、凝 膠、藥漿、藥棒、藥膏、乳膏、軟膏等。此外,這類的醫 59 201247612 藥組成物可選擇性的含有增溶劑、穩定劑、張力增強劑 (tonicity enhancing agent)、緩衝劑、或防腐劑。 於更一實施例中,化合物(即,本發明所述之環己烯酮 化合物)可以皮膚吸收方式投藥。於特定實施例中,皮廣吸 收劑型可為皮膚投藥裝置或皮膚投藥貼片,且可為油溶性 乳化劑、或溶解及/或分散在聚合物或黏著劑中之緩衝水性 溶液。於各種實施例中,這類的貼片係用以持續性、間隔 性、或依需求進行投藥。於其他實施例中,化合物(即,本 發明所述之環己稀萌化合物)之皮廣吸收方式投藥,可採用 離子電滲透貼片(iontophoretic patch)及其相似貼片。於特定 實施例中,皮膚吸收貼片係可控制傳送一化合物(即,本發 明所述之環己烯酮化合物)。於特定實施例中,減緩吸收速 率可藉由使用速率控制薄膜、或將化合物侷限在一聚合物 基質或膠艘中而達成。於另一實施例中,亦可使用吸收促 進劑以幫助吸收。吸收促進劑或載體可包括可吸收醫藥上 可接受之溶劑,其可幫助化合物穿過皮膚。例如,於一實 施例中’皮膚投藥裝置係為一繃帶形式,其包括:一背薄 膜;一用以容置化合物並選擇性容置載體之儲存空間;一 選擇性速率控制屏障,以依一控制且預定速率於長時間内 傳送藥物至主體的皮廣上;以及一安全元件,以確保裝置 對皮膚的安全性。 本發明所述之皮膚吸收劑型可使用本技術領域已知之 各種裝置進行投藥。裝置的例子可包括,但不限於美國專 利第 3,598,122 號、第 3,598,123 號、第 3,710,795 號、第 201247612 3,731,683號、第 3,742,951號、第 3,814,097號、第 3,921,636 號、第 3,972,995號、第 3,993,072號、第 3,993,073號、第 3,996,934號、第 4,〇31,894號、第 4,060,084號、第 4,〇69,307 號、第 4,077,407號、第 4,201,211號、第 4,230,105 號、第 4,292,299號、第 4,292,303號、第 5,334,168號、第 5,665,378 號、第 5,837,280號、第 5,869,090號、第 6,923,983號、第 6,929,801號、及第6,946,144號中所述之裝置。 本發明所述之皮廣吸收藥劑形式可包含本技術領域已 知之特定醫藥上可接受之賦形劑。於一實施例中’本發明 所述之皮濟吸收劑型係包括至少三種成分:(1)化合物藥劑 (即,本發明所述之環己烯酮化合物);P)吸收促進劑;以 及(3)水性佐劑。此外,皮膚吸收劑型可包括其他成分,例 如,但不限於:膠凝劑、乳霜或藥膏基質、及其相似成分。 於部分實施例中,皮膚吸收劑型更包括一織布或不織布底 墊材料,以幫助吸收並防止皮膚吸收劑型從皮膚上脫離》 於其他實施例中,本發明所述之皮膚吸收劑型係維持在飽 和或過度飽和狀態’以幫助藥劑其擴散至皮膚中。 於其他實施例中,化合物(即,本發明所述之環己烯酮 化合物)可製備成吸入式方式投藥。各種適合用於吸入式投 藥之劑型包括’但不限於喷霧、水氣或粉末形式。化合物 (即’本發明所述之環己烯酮化合物)之醫藥組成物一般係由 加壓裝置或喷霧器’並搭配使用一適當的推進劑(如,二氣 一氟曱烧、三氣一氟曱烷、二氣四氟乙烷、二氧化碳、或 其他適合氣體)’而形成霧狀喷霧形式以進行投藥。於特定 201247612 實施例中,加歷喷霧之單位劑量係透過閥決定’以傳送計 量供給之劑量。於特定實施例中’吸入器或吹藥器中使用 之明膠膠囊及儲存藥包(僅用以舉例用)可製備成含有一化 合物及適當粉末基質(如乳糖或殿粉)之粉末混合物。 鼻腔内投藥劑型為本技術領域已知劑型,且係如美國 專利公開第4,476,116號、第5,116,817號、及第6,391,452號 中所述,且每一專利係可併入本發明以供參考。根據上述 方法或其他本技術領域已知方法製備之藥劑,可製備成生 理食鹽水之溶液’其中’此藥劑係包括一化合物(即,本發 明所述之環己烯酮化合物)’且此溶液可包含本技術領域已 知之苯甲醇或其他適合的防腐劑、氟碳化合物、及/或其他 增溶劑或分散劑。例如,可參考Ansel,H.C. ei α/.Wherein each X and Y system is independently oxygen, NR5, or sulfur; R is hydrogen, or C(=0)C, -C8 alkyl; each R wide R2, and R3 are each independently hydrogen, Methyl or (CHdm-CHs; R4 is NR5R6, OR5, 〇C(=〇)R7, C(=0)0R5, C(=0)R5, halogen, 5 or 6 membered vinegar, Ci- Cg Hyun, C2_Cs dilute 'C2_C8 blocky' aryl, or glucosyl, wherein 5 or 6 membered ring lactone, C "C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, aryl, And one or more selected from the group consisting of NR5R6, 〇r5, oc(=o)r7, c(=o)or5, C(=0)R5, Cl-c8 alkyl, c2_c8, C2 Substituted by a substituent of -C8 alkynyl, CVC: 8 cycloalkyl, c丨-Cs haloalkyl, and c丨-C8 alkoxy; each R5, and R0 are each independently hydrogen, or €1 ( : 8 alkyl; R7 is (: 丨-C8 alkyl, 〇R5, or NR5R6; m is 1-12; and n is 1-12. In some embodiments, the nephropathy is glomerulosclerosis (eg, FSGS) In other embodiments, the nephropathy is renal glomerulonephritis (eg, immunoglobulin type A glomerulonephritis (igAN)). A class of steroid hormones that bind to the glucocorticoid receptor (GR). The term cortisol is derived from its regulation in glucose metabolism and its synthesis 49 201247612 in the adrenal cortex ( Adrenal cortex) and its steroid structure. Examples of auditory corticosteroids include, but are not limited to, cortisol (cortisol), cortisone acetate, prednisone, prednisone Prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fluorohydrogen acetate Fludrocortisone acetate, deoxycorticosterone acetate (DOCA), and aldosterone. Examples of drugs that act on immunones include, but are not limited to, cyclosporin, tac Tacrolimus, voclosporin and other calcineurin inhibitors, and rapamycin (siro) Limus) β Pharmaceutical Composition/Dosage Form The term "acceptable" means that the agent, composition, or active ingredient of the present invention does not cause a continuing adverse reaction to the daily life of the subject. Burdock is a fungus of the family Meripilaceae. The body of the burdock is generally flat or grows outward from the growth surface, and the sub-solid layer is oriented outward; in addition, the edges may be slightly raised and appear in a small bracket. Most of the species of Antrodia camphorata grow in the northern temperate forest and cause the brown rot of trees. In addition, some of the special species of this fungus are more effective, and are often used as one of the traditional Chinese medicines in Taiwan. As used herein, the term "carrier" refers to a relatively non-toxic compound or chemical that aids in the delivery of a compound to a cell or tissue. 50 201247612 In the present invention, the term "co-administered" includes the administration of a selected therapeutic agent to a patient, and further includes a method of treating the agents in the same or different administration manners, or at the same or different administration times. The term "diluent" means a compound used to dilute the compound used prior to administration. Diluents can also be used to stabilize compounds because they provide a more stable environment. Salt buffer solutions commonly used in the art (which can be used to control or maintain pH) can also be used as diluents including, but not limited to, citrate buffer solutions. In the present invention, the term "effective dose" or "therapeutically effective dose" means an appropriate dose of the agent or compound which, to some extent, alleviates one or more conditions of the disease or condition being treated. In this way, the symptoms, symptoms, or causes of the disease can be alleviated and/or alleviated, or other changes can be made to the physiological system according to the needs. The "effective dose" used in the treatment of the disease means that the disease can be clinically provided. Significantly improved compositional doses required 'where the composition is a system comprising the compounds disclosed herein. In addition, appropriate "effective" doses are determined according to methods such as dose escalation tests, under different conditions. In the present invention, the term "promotional enhancement" refers to the effect or duration of an increase or extension of a predetermined effect. ^ Bellow, therefore, the term "promotion" or "擗%^„9" refers to the dragon that increases or prolongs the effectiveness or duration, or the line. Other therapeutic agents in the treatment can achieve an effect of increasing or prolonging. Further, "enhanced effective dose" as used in the present invention means an appropriate dose which can enhance the effect of another therapeutic agent in a predetermined system. 5] 201247612 In the present invention, the term "metabolite" refers to a compound derivative formed by metabolism of a compound. The term "active metabolite" refers to a physiologically active compound derived from the metabolism of a compound. Things. Further, in the present invention, the term "metabolism" refers to all processes in which a specific component is changed by an organism (including, but not limited to, hydrolysis reaction and enzyme catalytic reaction). Therefore, 'enzymes can make special structural changes to a compound. For example, cytochrome Ρ450 can catalyze a variety of oxidation and reduction reactions, while uridine diphosphate glucuronyltransferase catalyzes the transfer of active glucuronic acid molecules to aromatic alcohols, fatty alcohols, Carboxylic acids, amines, and sulphydryl radicals are trapped. In the present invention, the metabolite of the compound is also selectively permeable: it is defined by administering the compound to a host and analyzing the tissue sample of the host, or culturing the compound with liver cells in vitro and analyzing the resulting compound. In the present invention, the term "pharmaceutical combination" means a product formed by mixing or combining more than one active ingredient, and comprises both fixed and non-fixed compositions of the active ingredient. Here, the term "fixed composition" means that the active ingredient (a compound such as the cyclohexanone compound of the present invention) and an auxiliary agent are simultaneously administered to a patient in a single object or dosage form. The term "non-fixed composition" means that the active ingredient (a compound such as the cyclohexenone compound of the present invention) and an adjuvant are administered in separate dosage forms simultaneously, together, or without specific time intervals. A patient in which such a mode of administration provides an effective amount of two compounds 52 201247612 to a patient. In addition, non-fixed compositions can also be used in cocktail therapy, such as by administering three or more active ingredients. The term "pharmaceutical composition" means a mixture of a compound (ie, a cyclohexenone compound of the present invention) and other chemical components, such as a carrier, a stabilizer, a diluent, a dispersant. , suspending agents, thickeners, and/or excipients. Pharmaceutical compositions can help a compound be administered to a species. Various techniques for administering compounds known in the art to which the present invention pertains include, but are not limited to, intravenous, oral, spray administration, parenteral administration (non-digestive administration), ocular administration, pulmonary administration (inhalation administration). And external use of drugs. In addition, the term "subject" or "patient" includes mammals; examples of mammals include, but are not limited to, any species of the lactation network: humans, non-human primates such as orangutans and other apes and monkey species; farms Animals such as cattle, horses, sheep, goats, pigs; livestock such as rabbits, dogs, and cats; experimental animals including scorpion animals such as rats, mice, guinea pigs, and other similar animals. In one embodiment, the mammalian system is a human. In the present invention, the term "treatment" includes: preventing or inhibiting other diseases by inhibiting and/or treating at least one condition for alleviating, alleviating or ameliorating a disease or symptom (for example, preventing the occurrence of a disease or symptom), Alleviate the symptoms or symptoms that cause (4) or symptoms to be caused by symptoms or symptoms, or to inhibit the disease or symptoms. The route of administration is not limited to: oral administration, ocular, lung, mucosa, and the administration routes suitable for the present invention include intravenous injection, intestinal tract, spray type, parenteral, 201247612 skin surface, vagina, ear, nasal cavity, And external use of drugs. In addition, examples of parenteral administration include intramuscular, subcutaneous, intravenous, intrathecal injections, and also include intracerebral meninges, direct intraperitoneal, intraperitoneal intralymphatic, intranasal injections. In a particular embodiment, the compounds described herein are generally prepared as a depot or sustained release dosage form' and are administered in a local rather than systemic manner, e.g., by injecting the compound directly into an organ. In a particular embodiment, a long-acting dosage form of the drug can be administered by means of colonization (e.g., subcutaneous or intramuscular) or intramuscular injection. Further, in other embodiments, the drug can be administered by a standard dry drug delivery system (e.g., a liposome coated with an organ-specific anti-Zhao). In this embodiment, the liposomes can be directed to an organ and selectively absorbed by the organ. In other embodiments, the compounds of the invention may be prepared in a fast-release dosage form, a slow release dosage form, or an immediate release dosage form. In other embodiments, the compounds of the invention may be administered in a topical manner. Pharmaceutical Composition/Life [Type In some embodiments, the compounds of the present invention can be prepared as a pharmaceutical composition. In a particular embodiment, the pharmaceutical composition can be prepared in a conventional manner using one or more physiologically acceptable carriers; and the physiologically acceptable carrier includes excipients and adjuvants which aid in the processing of the active compound The process is prepared to produce a pharmaceutically acceptable agent. The appropriate dosage form is chosen according to the route of administration. Any pharmaceutically acceptable technique, carrier, and excipient can be used to prepare the pharmaceutical compositions of the present invention: Remington. The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 54 201247612 1995); Hoover, John E., Remington's Pharmaceutical •Scz.ewce·?,Mack Publishing Co., Easton,Pennsylvania 1975; Liberman, HA and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York , NY, 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkinsl 999). The present invention provides a pharmaceutical composition comprising a compound (e.g., a cyclohexenone compound of the present invention) and a pharmaceutically acceptable diluent, excipient, or carrier. In a particular embodiment, The compound can be administered by a combination of a compound (e.g., a cyclohexenone compound of the present invention) and a pharmaceutical composition of another active ingredient in a combined therapeutic manner. The present invention includes all compositions of the present invention relating to the active ingredients described in the paragraphs of the Combination Therapy. In a particular embodiment, the pharmaceutical composition comprises one or more compounds (e.g., a cyclohexenone compound of the invention). The pharmaceutical composition of the present invention refers to a mixture of a compound (e.g., a cyclohexenone compound of the present invention) and other compounds, such as a carrier, a stabilizer, a diluent, a dispersing agent, a suspending agent, and a thickening agent. And/or excipients. In a particular embodiment, the pharmaceutical composition can facilitate administration of the compound to an organism. In some embodiments, the therapeutic method of the present invention is to administer a therapeutically effective amount of a compound (eg, a cyclohexenone compound of the present invention) as a pharmaceutical composition to a subject having a disease or symptom. . In a particular embodiment, the mammalian system is a human. In a particular embodiment, the therapeutically effective dose will vary depending on the severity of the disease, the age of the subject and the extent of the health, the potency of the compound, and other factors. Furthermore, the compounds described in the present invention can be used alone or in combination with one or more therapeutic agents (as a component of a mixture). In one embodiment, the compound (e.g., the cyclohexenone compound of the present invention) is prepared in the form of an aqueous solution. In certain embodiments, examples of aqueous solutions may be selected from water-soluble, physiologically compatible buffers (such as Hank's solution, Ringer's solution, or physiological saline). In the embodiments, the compound (e.g., the cyclohexenone compound of the present invention) can be prepared into a mucosal dosage form. In a specific embodiment, the mucosal administration form includes a penetrant that can properly penetrate the mucosal barrier. In other embodiments, the compounds described herein are prepared in other parenteral dosage forms and suitable dosage forms include aqueous or non-aqueous solutions. In particular embodiments, such solutions include physiologically compatible buffers and / or an excipient. In another embodiment, the compound of the present invention can be prepared into an oral dosage form. Here, the compound of the present invention (i.e., the cyclohexenone compound of the present invention) is contained. The preparation is carried out by mixing the active compound with, for example, a pharmaceutically acceptable compound or an excipient. In various embodiments, the compound of the present invention can be prepared into an oral dosage form. Examples include lozenges, powders, tablets, pills, capsules, syrups, gels, medicated syrups, elixirs, steroids, suspensions, and the like. In a particular embodiment, the oral dosage form can be one or more solids. The excipient is mixed with one or more of the compounds of the present invention, and the resulting mixture is selectively milled, and if necessary, the powder mixture can be processed to obtain a tablet or pill core after addition of a suitable adjuvant. Suitable excipients are, in particular, fillers, such as sugars containing lactose, sucrose, mannitol, 56 201247612 or sorbitol; cellulose preparations such as maize starch, wheat flour, rice starch, potato starch, gelatin , rubber gum, sulfhydryl cellulose, microcrystalline cellulose, hydroxypropyl decyl cellulose, sodium carboxymethyl cellulose; or other such as polyvinylpyrrolidine (PVP or povidone) or calcium phosphate beta In the examples, 'disintegrator may be optionally added. Examples of the disintegrant may include: Croscarmellose sodium, polyethylene bismuthone, agar Or alginate or a salt thereof (such as sodium alginate). In one embodiment, the agent such as the core of the pill and the lozenge may be one or more suitable coating layers. In a specific embodiment, the concentrated sugar solution is used. To encapsulate the agent. The saccharide solution may optionally include other additional ingredients such as gum arabic, talc, polyvinylpyrrolidone carbopol gel, polyethylene glycol, and/or titanium dioxide; a coating solution; and a suitable organic solvent or solvent mixture β dye and/or pigment may also be selectively added to the coating for identification purposes. In addition, dyes and/or pigments may also be selectively used to A composition for labeling different active compound agents. In a particular embodiment, a therapeutically effective amount of at least one compound of the invention can be prepared in a form other than an oral dosage form. The oral dosage form comprises a push-fit capsule made of gelatin (10), a sputum, and a soft and sealed capsule made of gelatin and a plasticizer such as glycerol or sorbitol. In a particular embodiment, the push-fit capsule comprises an active ingredient in admixture with one or more fillers. Examples of the filler may include lactose, an adhesive such as starch, and/or a lubricant such as talc or magnesium stearate, and optionally include a stabilizer. In other embodiments, the soft capsules comprise an active compound dissolved or suspended in a suitable liquid in the form of 57 201247612 or above. Examples of suitable liquids include: one or more fatty oils, liquid paraffin, or liquid polyethylene glycol. Further, a stabilizer may be optionally added. In other embodiments, a therapeutically effective dose of at least one compound of the invention can be prepared in the form of a buccal dosage form or a sublingual dosage form. Examples of suitable dosage forms for oral or sublingual administration include lozenges, tablets, or gels. In still other embodiments, the compounds of the invention may be prepared as parenteral injection forms' which include dosage forms suitable for bolus injection or continuous infusion. In a particular embodiment, the injectable dosage form is in unit dosage form (e.g., in the form of an ampoule), or in a multi-dose package. In addition, a preservative can be optionally added to the injectable dosage form. In still other embodiments, the pharmaceutical composition of the compound (ie, the cyclohexenone compound of the present invention) is dissolved in an oily or aqueous vehicle to prepare a sterile suspension, solution, or emulsifier. A dosage form for parenteral injection. Parenteral injection forms may optionally include such agents as suspending, stabilizing, and/or dispersing agents. In a particular embodiment, the pharmaceutical dosage form for parenteral administration comprises an aqueous solution of the active compound in a water soluble form. In other embodiments, the suspension of the active compound is prepared as a suitable oily injection suspension, as an example of a suitable oil-soluble solvent or vehicle for use in the pharmaceutical compositions of the invention, and may include a fatty oil such as sesame oil, or For example, ethyi eleate or a synthetic fatty acid ester of triglyceride, or a liposome. In a particular embodiment, an aqueous injection suspension contains a substance that increases the viscosity of the suspension, such as carboxy hydrazine. Cellulose sodium, sorbitol, or dextran. In addition, the suspension optionally includes suitable stabilizers or agents to enhance the dissolution of the compound to provide a highly concentrated solution. Alternatively, in other embodiments, the active ingredient may be in powder form for mixing with a suitable vehicle (e.g., sterile pyrogen-free water) prior to use. In one embodiment, the compound (i.e., the cyclohexenone compound of the present invention) is prepared as a solution for parenteral injection by the method of the present invention or by methods known in the art, and an autoinjector is used. Dosing. Automated injections are disclosed in U.S. Patent Nos. 4,031,893, 5,358,489, 5, 540, 664, 5, 665, s. 71, 5, 695, 472, and WO/2005/087297, each of which is incorporated herein by reference. The contents are incorporated herein by reference. In general, all autoinjectors include a solution of hips to be injected, the solution comprising a compound (ie, the cyclohexenone compound of the present invention. In addition, the autoinjector system includes: one for containing a solution The storage space 'this storage space is fluidly connected to a needle for administration; and the automatic insertion needle mechanism for inserting the needle onto a patient to deliver the medicament to the patient. For example, the syringe can be supplied for about 0.3. mL, 0.6 mL, 1.0 mL, or other suitable volume of solution, and the mother 1 ml of the solution contains a compound having a concentration of about 5 mg to 50 mg (i.e., the cyclohexenone compound of the present invention). Each of the syringes may deliver only one dose of the compound. In still other embodiments, the compound (ie, the cyclohexanone compound of the present invention) may be administered externally. Here, the compounds of the present invention can be prepared into various types. Compositions which can be administered externally, such as solutions, suspensions, emulsions, gels, slurries, sticks, ointments, creams, ointments, etc. In addition, such a medicine 59 201247612 pharmaceutical composition Optionally, it contains a solubilizing agent, a stabilizer, a tonicity enhancing agent, a buffering agent, or a preservative. In a further embodiment, the compound (ie, the cyclohexenone compound of the present invention) can be skin In a specific embodiment, the dermal absorption dosage form may be a skin administration device or a skin administration patch, and may be an oil-soluble emulsifier, or a buffered aqueous solution dissolved and/or dispersed in a polymer or an adhesive. In various embodiments, such patches are used for sustained, intermittent, or on demand. In other embodiments, the skin of the compound (ie, the cycloheximide compound of the present invention) For the broad absorption mode of administration, an iontophoretic patch and similar patches can be used. In a particular embodiment, the skin-absorbing patch can control the delivery of a compound (ie, the cyclohexenone of the present invention) Compounds. In certain embodiments, slowing down the rate of absorption can be achieved by using a rate controlling film, or by confining the compound to a polymer matrix or gelatinous vessel. In one embodiment, an absorption enhancer can also be used to aid absorption. The absorption enhancer or carrier can include a pharmaceutically acceptable solvent that can aid in the passage of the compound through the skin. For example, in one embodiment, a 'skin delivery device The invention is in the form of a bandage comprising: a back film; a storage space for accommodating the compound and selectively accommodating the carrier; and a selective rate control barrier for delivering the drug to the drug for a long time at a controlled and predetermined rate to The body of the body is widened; and a security element to ensure the safety of the device to the skin. The skin absorbent dosage form of the present invention can be administered using a variety of devices known in the art. Examples of devices can include, but are not limited to, the United States. Patent Nos. 3,598,122, 3,598,123, 3,710,795, 201247612 3,731,683, 3,742,951, 3,814,097, 3,921,636, 3,972,995, 3,993,072, 3,993,073, 3,996,934, 4, 〇31,894, 4,060,084, 4, 〇69,307, 4,077,407, 4,201,211 No. 4,230,105, No. 4,292,299, No. 4,292,303, No. 5,334,168, No. 5,665,378, No. 5,837,280, No. 5,869,090, No. 6,923,983, No. 6,929,801, No. 6,946,144 and second of said apparatus. The broadly absorbing dosage forms of the present invention may comprise specific pharmaceutically acceptable excipients known in the art. In one embodiment, the skin-absorbing dosage form of the present invention comprises at least three components: (1) a compound agent (ie, a cyclohexenone compound of the present invention); P) an absorption enhancer; and (3) ) aqueous adjuvant. In addition, the skin absorbent dosage form can include other ingredients such as, but not limited to, a gelling agent, a cream or ointment base, and the like. In some embodiments, the skin absorbent dosage form further comprises a woven or non-woven backing pad material to aid in absorbing and preventing detachment of the skin absorbent dosage form from the skin. In other embodiments, the skin absorbent dosage form of the present invention is maintained Saturated or oversaturated state' to help the agent spread into the skin. In other embodiments, the compound (i.e., the cyclohexenone compound of the present invention) can be prepared for administration by inhalation. A variety of dosage forms suitable for inhaled administration include, but are not limited to, spray, moisture or powder form. The pharmaceutical composition of the compound (ie, the cyclohexenone compound of the present invention) is generally a pressurizing device or a sprayer' and is used in combination with a suitable propellant (eg, digas-fluorine, trigas) The monofluorosilane, dioxane tetrafluoroethane, carbon dioxide, or other suitable gas) form a mist spray for administration. In the specific 201247612 embodiment, the unit dose of the calendar spray is determined by the valve to deliver the dose supplied by the meter. Gelatin capsules and storage packs (for illustrative purposes only) used in the "inhaler or insufflator" in a particular embodiment can be prepared as a powder mixture containing a compound and a suitable powder base such as lactose or powder. The intranasal administration form is a dosage form known in the art and is described in U.S. Patent Nos. 4,476,116, 5,116,817, and 6,391,452, each of which is incorporated herein by reference. . A solution of physiological saline can be prepared according to the above method or other agents prepared by methods known in the art, wherein the agent comprises a compound (i.e., the cyclohexenone compound of the present invention) and the solution Benzyl alcohol or other suitable preservatives, fluorocarbons, and/or other solubilizing or dispersing agents known in the art may be included. For example, refer to Ansel, H.C. ei α/.

Pharmaceutical Dosage Forms and Drug Delivery System, Sixth Ed. (1995)»較佳為,此些組成物及藥劑可與適當的 無毒醫藥上可接受之成分一同製備。此些成分可參考本技 術中常用之參考資料,如REMINGTON: THE SCIENCE AND PRACTICE PHARMACY, 21st edition, 2005。此外,載體可 根據所需求之鼻腔内藥劑劑型特性做適當的選擇,如:溶 液、懸浮液、藥膏、或凝膠等。鼻腔内投藥劑型一般係含 有大量的水以及活性成分。此外,亦可包含少量其他成分, 如pH調整劑、乳化劑或分散劑、防腐劑、介面活性劑、凝 膠劑、或緩衝劑及其他穩定劑與增溶劑。較佳為,鼻腔内 投藥劑型為與鼻内分泌物具有等滲透壓。 當以吸入式方式投藥時,本發明所述之化合物可製備 成喷霧、水氣或粉末。本發明所述之醫藥組成物一般係由 62 201247612 加壓裝置或喷霧器,並搭配使用一適當的推進劑(如,二氣 二氟甲烷、三氣一氟曱烷、二氯四氟乙烷'二氧化碳、或 其他適合氣體),而形成霧狀喷霧形式以進行投藥。於加壓 喷霧之例子中,單位劑量係透過閥決定,以傳送計量供給 之劑量。此外,吸入器或吹藥器中使用之明膠膠囊及儲存 藥包(僅用以舉例用)可製備成含有本發明之化合物及適當 粉末基質(如乳糖或澱粉)之粉末混合物。 於再其他實施例中,化合物(即,本發明所述之環己烯 酮化合物)可製備成直腸用組成物,如灌腸劑、直腸凝膠、 直腸泡沫劑、直腸喷霧劑、栓劑、膠體栓劑、或留置灌腸 劑’其包含習知栓劑基質(如可可油或其他甘油酯)、以及合 成聚合物(如聚乙烯吡咯烷酮、PEG及其相似物)。此外,於 組成物之栓劑劑型中’可將可可油選擇性的與低熔點堪(例 如’但不限於脂肪酸及甘油酯混合物)使用,且低溶點壤會 先融化。 於特定實施例中,醫藥組成物係與一種或以上生理上 可接受之載體’透過任何習知方式製備,其中,生理上可 接受之載體包括賦形劑及輔劑,其可幫助活性化合物之製 備並適用於醫藥上》此外,可依照所選擇之投藥路徑來選 擇適當的劑形。若適用且符合本技術領域已知之知識,可 選擇性的使用任何醫藥上可接受之技術、載體、及賦形劑。 包含化合物(即,本發明所述之環己烯酮化合物)之醫藥組成 物可以習知之方法製備,例如(僅用以舉例用)習知之混合、 63 201247612 溶解、研磨、藥丸製作、粉末化、乳化、製成膠囊'包覆 或壓錠等製程。 醫藥组成物可包括:至少一醫藥上可接受之載體、稀 釋劑或賦形劑;以及至少一本發明所述之化合物(即,本發 明所述之環己烯酮化合物),此化合物係作為一活性成分。 活性成分係為游離酸或游離驗形式,或為醫藥上可接受之 鹽類形式。此外,本發明所述之方法及醫藥組成物係包括 晶體型態(即多晶型物)’亦包括此些化合物之具有相同活性 之活性代謝物。本發明所述之化合物之互變異構體亦併入 本發明所述化合物之範圍中。此外,本發明所述之化合物 亦可包含非溶劑形式、以及溶劑形式,其中溶劑形式係使 用醫藥上可接受之溶劑,如水、乙醇、及相似溶劑。於本 發明中,化合物之溶劑形式亦併入本發明所揭示之範圍 中。此外,醫藥組成物可選擇性的包括其他藥物或藥劑、 載體、佐剤(如保存劑、穩定劑、加濕劑、或乳化劑)、溶液 促進劑、用以調整滲透壓之鹽類、緩衝液、及/或其他治療 上有效之物質。 於本發明中,含有本發明化合物之組成物之製備方法 係包括·將〖合物與一種或以上低活性且醫藥上可接受之 賦形劑或載趙-同§己製,而製備成固態半固態或液態之 形式。固態組成物包括,但不限於:粉末、錢劑、分散顆 粒、膠囊、及栓劑。液態組成物包括:溶有化合物之溶液; 含有化合物之乳液;或含有微脂體、膠質粒子、或奈米粒 子之溶液’其中微脂體、„粒子、或奈米粒子内係容置 64 201247612 有本發明所述之化合物。半m成物包括,但不限於·· 凝膠、懸净液、或乳霜。本發明所述之醫藥組成物之型態 可包括:㈣溶液或懸浮液、可於使用前於液趙中形成溶 液或懸我之固態形式、或者是乳液。這些組成物亦可選 擇1±的含有}量無毒之輔劑,如加濕劑或乳化劑、pH緩衝 劑等輔劑。 於部分實施例中,本發明中所述之包含至少一化合物 (即,本發明所述之環己烯酮化合物)之醫藥組成物,其係為 一液態形式,其中藥劑係在溶液中、懸浮液中、或兩者中。 一般而言,當組成物係以溶液或懸浮液的方式投藥時藥 劑之第部分係為溶液形式,而藥劑之第二部分則具有特 殊形式,如於液態基質中之懸浮液。於部分實施例中液 態組成物包括凝膠劑型。於其他實施例中,液態組成物係 為水溶液形式。 於特定實施例中,醫藥水性懸浮液包括一種或以上之 聚合物,以作為懸浮劑》聚合物可包括如纖維素聚合物之 水溶性聚合物(如:羥丙基甲基纖維素)、以及如交聯含羧基 聚合物之非水溶性聚合物。本發明所述之特定醫藥組成物 包括黏膜枯附聚合物(mucoadhesive polymer),其可選自由 如:叛甲基纖維素、卡波姆(carbomer)(丙稀酸聚合物)、聚(甲 基丙烯酸甲酯)、聚丙烯胺、聚卡波非(p〇lycarb〇phil)、丙烯 酸/丙稀酸丁醋共聚物、海藻酸納(sodium alginate)、及葡萄 聚糖。 65 201247612 醫藥組成物亦選擇性的包含增溶劑,以幫助化合物 (即,本發明所述之環己烯酮化合物)之溶解度。「增溶劑」 一詞一般係包括:一可用以形成膠束溶液(micellar solution) 之試劑、或一溶液試劑。此外,如聚山梨醇S旨80 (polysorbate 80)之特定可接受之非離子界面活性劑亦可用以作為增溶 劑,且增溶劑亦可為眼部可接受之二醇、聚二醇(如,聚乙 二醇400)、及二醇醚。 再者,醫藥組成物可選擇性的包括一種或以上pH調整 劑或緩衝劑。其中,pH調整劑或緩衝劑包括:如乙酸、硼 酸、檸檬酸、乳酸、磷酸、氫氣酸等酸類;如氫氧化鈉、 磷酸鈉、硼酸鈉、檸檬酸鈉、醋酸鈉、乳酸鈉、三-羥基甲 基氨基甲烧(tris-hydroxymethylaminomethane)等驗類;以及 如檸檬酸酯/葡萄糖(dextrose)、碳酸氫鈉、氣化銨等緩衝物 質。這類的酸、驗、及緩衝物質之含量,係為一可維持組 成物pH值之可接受範圍。 此外,醫藥組成物可選擇性的包括一種或以上鹽類, 此鹽類所需的量係用以將組成物之滲透壓維持在一可接受 的範圍。可使用之鹽類包括:鈉、鉀、或銨根之陽離子; 以及氣、檸檬酸根、抗壞血酸根(ascorbate)、硼酸根、磷酸 根、碳酸氫根(bicarbonate)、硫酸根、硫代硫酸根、及亞硫 酸根之陰離子。此外,適當的鹽類例子包括:氣化鈉、氣 化鉀、硫代硫酸鈉、亞硫酸鈉、及硫酸銨。 其他醫藥組成物可選擇性的包括一種或以上防腐劑, 以抑制細菌活性。適當的防腐劑包括:如硼酸苯汞(merfen) 66 201247612 及硫柳汞(thiomersal)之含汞物質;穩定之二氧化氣 (chlorine dioxide),及如笨紫氣敍(benzaik〇nium chloride)、 十六烧基二曱基 >臭化链(cetyltrimethylammonium bromide)、及氯化十六烷吡啶(cetyipyridinium chloride)之四 級敍鹽化合物。 於其他醫藥組成物中,可包括一種或以上界面活性 劑,以增強物理穩定性或其他特定。適合的非離子界面活 性劑包括:聚氧6稀脂肪睃甘油醋(polyoxyethylene fatty acid glyceride)及植物油,如:聚氧乙稀(60)氫化乾麻油; 以及聚氧乙烯烧基醚(polyoxyethylene alkylethers)及炫•基 苯基喊,如辛基紛聚謎10(〇ctoxynol 10)、辛基紛聚鍵40。 於再其他組成物中,可包含一種或以上抗氧化劑’以 增加所需之化學穩定性。適合之抗氧化劑包括(僅用於示 例)··抗壞血酸及偏亞硫酸氫納(sodium metabisulfite)。 於特定實施例中,醫藥水性懸浮液組成物係包裝成單 一劑量且不可重複開啟之容器中。此外’亦可使用多劑量 可重複開啟之容器,在此,組成物中一般係包含防腐劑。 於另一實施例中,亦可使用用於疏水性醫藥化合物之 投藥系統。例如,本發明可使用微脂體及乳化劑作為投藥 之媒介或載體。於特定實施例中,可使用如N-曱基吡咯烷 酮之有機溶劑。於另外實施例中,本發明所述之化合物可 使用持續釋放系統進行投藥,如含有治療藥劑之固態疏水 聚合物之半滲透基質。此外,各種持續釋放材料亦可用於 本發明中。於部分實施例中,持續釋放膠囊可於數小時, 67 201247612 甚至超過24小時釋放化合物。隨著治療藥劑之化學特性及 生物穩定性之不同,可額外添加蛋白質穩定劑。 於特定實施例中,本發明所述之藥劑可包括一種或以 上抗氧化劑、金屬螯合劑、含硫醇化合物、及/或其他一般 常用之穩定劑。這類穩定劑的例子包括,但不限於:(a)約 0.5%至約2% w/v之甘油、(b)約0.1%至約1% w/v之蛋胺酸 (methionine)、(c)約0.1 %至約2% w/v之單硫代甘油 (monothioglycerol)、(d)約 ImM至約 10mM之 EDTA、(e)約 0.01 % 至約 2% w/v 之抗壞血酸、(f) 0.003% 至約 0.02% w/v 之聚山梨酸酯 80(p〇lysorbate 80) 、(g) 0.001。/。至約 0.05% w/v之聚山梨酸醋20、(h)精胺酸(arginine)、⑴肝素 (heparin)、⑴硫酸葡聚糖(dextran sulfate) 、(k)環糊精 (cyclodextin)、(1)戍聚糖聚硫酸醋(pentosan polysulfate)及 其他類肝素(heparinoid)、(m)如鎂離子及鋅離子之二價陽 離子;或(η)其混合物。 【實施方式】 實施例1:示範環己烯酮化合物之製備 示範化合物1(即,Antroq)係由牛樟芝之固態發酵菌絲 趙,以習知方法分離而得(Lee,et al.,Med, 73: 1412-1415, 2007)。根據先前研究及使用經驗,化合物1之有 效劑量為 10-50 mg/kg 體重(Chang, et al.,Preferably, such compositions and agents are prepared with suitable non-toxic pharmaceutically acceptable ingredients. These components can be referred to reference materials commonly used in the art, such as REMINGTON: THE SCIENCE AND PRACTICE PHARMACY, 21st edition, 2005. In addition, the carrier may be suitably selected depending on the desired formulation of the dosage form in the nasal cavity, such as a solution, suspension, ointment, or gel. Intranasal administration usually contains a large amount of water and active ingredients. In addition, minor amounts of other ingredients such as pH adjusting agents, emulsifiers or dispersing agents, preservatives, surfactants, gelling agents, or buffering agents, and other stabilizers and solubilizing agents may be included. Preferably, the intranasal administration mode has an isotonic pressure with the nasal secretions. When administered by inhalation, the compounds of the invention may be prepared as a spray, moisture or powder. The pharmaceutical composition of the present invention is generally a 62 201247612 pressurizing device or a sprayer, and is used in combination with a suitable propellant (eg, di-halogenated difluoromethane, tri-gas-fluorodecane, dichlorotetrafluoroethylene). The alkane 'carbon dioxide, or other suitable gas), forms a mist spray for administration. In the case of a pressurized spray, the unit dose is determined by a valve to deliver a metered dose. In addition, gelatin capsules and storage packs (for example only) used in inhalers or insufflators can be prepared as a powder mixture containing a compound of the present invention and a suitable powder base such as lactose or starch. In still other embodiments, the compound (ie, the cyclohexenone compound of the present invention) can be prepared into a rectal composition such as an enema, a rectal gel, a rectal foam, a rectal spray, a suppository, or a colloid. A suppository, or a retention enemas, which comprise a conventional suppository base (such as cocoa butter or other glycerides), and synthetic polymers (such as polyvinylpyrrolidone, PEG, and the like). Further, in the suppository dosage form of the composition, the cocoa butter can be selectively used with a low melting point (e.g., but not limited to a mixture of a fatty acid and a glyceride), and the low solubility point first melts. In a particular embodiment, the pharmaceutical composition is prepared by any conventional means with one or more physiologically acceptable carriers, wherein the physiologically acceptable carrier includes excipients and adjuvants which aid in the active compound. It is prepared and applied to medicine. In addition, an appropriate dosage form can be selected according to the selected route of administration. Any pharmaceutically acceptable technique, carrier, and excipient can be optionally employed, where applicable and in accordance with what is known in the art. A pharmaceutical composition comprising a compound (i.e., a cyclohexenone compound of the present invention) can be prepared by a conventional method, for example, by way of example only, a conventional mixture, 63 201247612 dissolution, grinding, pelleting, powdering, Emulsified, made into capsules, coated or pressed. The pharmaceutical composition may comprise: at least one pharmaceutically acceptable carrier, diluent or excipient; and at least one compound of the invention (ie, a cyclohexenone compound of the invention) which is An active ingredient. The active ingredient is in free acid or free form or in the form of a pharmaceutically acceptable salt. Furthermore, the methods and pharmaceutical compositions of the present invention include crystalline forms (i.e., polymorphs) and also include active metabolites of such compounds having the same activity. Tautomers of the compounds described herein are also included within the scope of the compounds described herein. Further, the compounds of the present invention may also be in a non-solvent form, as well as a solvent form, wherein the solvent form is a pharmaceutically acceptable solvent such as water, ethanol, and the like. In the present invention, the solvent form of the compound is also included in the scope of the present invention. In addition, the pharmaceutical composition may optionally include other drugs or agents, carriers, adjuvants (such as preservatives, stabilizers, moisturizers, or emulsifiers), solution accelerators, salts for adjusting osmotic pressure, and buffers. Liquid, and / or other therapeutically effective substances. In the present invention, the preparation method of the composition containing the compound of the present invention comprises: preparing the compound into a solid state with one or more low-activity and pharmaceutically acceptable excipients or Semi-solid or liquid form. Solid compositions include, but are not limited to, powders, money, dispersed granules, capsules, and suppositories. The liquid composition includes: a solution in which a compound is dissolved; an emulsion containing a compound; or a solution containing a liposome, a colloidal particle, or a nanoparticle, wherein the microlipid, „particle, or nanoparticle internal system is accommodated 64 201247612 There is a compound according to the present invention. The semi-m composition includes, but is not limited to, a gel, a suspension, or a cream. The form of the pharmaceutical composition of the present invention may include: (4) a solution or suspension, It can be used to form a solution or suspend the solid form or emulsion in liquid Zhao before use. These compositions can also be selected as 1± containing non-toxic auxiliary agents such as humidifying agents or emulsifiers, pH buffering agents, etc. In some embodiments, the pharmaceutical composition of the present invention comprising at least one compound (ie, the cyclohexenone compound of the present invention) is in a liquid form, wherein the agent is in solution In the middle, in the suspension, or both. In general, when the composition is administered as a solution or suspension, the first part of the agent is in the form of a solution, and the second part of the agent has a special form, such as In liquid matrix The liquid composition in some embodiments includes a gel dosage form. In other embodiments, the liquid composition is in the form of an aqueous solution. In a particular embodiment, the pharmaceutical aqueous suspension comprises one or more polymers, The polymer as a suspending agent may include a water-soluble polymer such as a cellulose polymer (e.g., hydroxypropylmethylcellulose), and a water-insoluble polymer such as a crosslinked carboxyl group-containing polymer. Specific pharmaceutical compositions include mucoadhesive polymers, such as: methyl cellulose, carbomer (acrylic acid polymer), poly(methyl methacrylate), Polyacrylamide, polycarbophil (p〇lycarb〇phil), acrylic acid/acrylic acid butyl vinegar copolymer, sodium alginate, and glucomannan. 65 201247612 Pharmaceutical compositions are also selectively included a solvent to aid in the solubility of the compound (i.e., the cyclohexenone compound of the present invention). The term "solubilizer" generally includes: one can be used to form a micellar solution. Reagent, or a reagent solution. In addition, a specific acceptable nonionic surfactant such as polysorbate 80 can also be used as a solubilizing agent, and the solubilizing agent can also be an ocularly acceptable diol or polyglycol (eg, Polyethylene glycol 400), and glycol ethers. Further, the pharmaceutical composition may optionally include one or more pH adjusters or buffers. Wherein, the pH adjusting agent or buffering agent comprises: an acid such as acetic acid, boric acid, citric acid, lactic acid, phosphoric acid, hydrogen acid; such as sodium hydroxide, sodium phosphate, sodium borate, sodium citrate, sodium acetate, sodium lactate, tris-hydroxyl Tris-hydroxymethylaminomethane and the like; and buffer substances such as citrate/dextrose, sodium bicarbonate, and ammonium hydride. The amount of such acid, test, and buffer materials is an acceptable range to maintain the pH of the composition. Further, the pharmaceutical composition may optionally include one or more salts which are required to maintain the osmotic pressure of the composition within an acceptable range. Salts which may be used include: sodium, potassium, or ammonium cations; and gas, citrate, ascorbate, borate, phosphate, bicarbonate, sulfate, thiosulfate, And anions of sulfite. Further, examples of suitable salts include: sodium gasification, potassium hydride, sodium thiosulfate, sodium sulfite, and ammonium sulfate. Other pharmaceutical compositions may optionally include one or more preservatives to inhibit bacterial activity. Suitable preservatives include: mercury-containing substances such as merfen 66 201247612 and thiomersal; stable chlorine dioxide, and benzaik〇nium chloride, sixteen a quaternary salt compound of cetyltrimethylammonium bromide and cetidipyridinium chloride. In other pharmaceutical compositions, one or more surfactants may be included to enhance physical stability or other specificity. Suitable nonionic surfactants include: polyoxyethylene fatty acid glyceride and vegetable oils such as polyoxyethylene (60) hydrogenated dry sesame oil; and polyoxyethylene alkylethers And Hyun • base phenyl shout, such as octyl gathering 10 (〇 ctoxynol 10), octyl condensed key 40. In still other compositions, one or more antioxidants may be included to increase the desired chemical stability. Suitable antioxidants include (for example only) ascorbic acid and sodium metabisulfite. In a particular embodiment, the pharmaceutical aqueous suspension composition is packaged in a single dose and non-repeatable opening container. Further, a multi-dose re-openable container may be used, where the composition generally contains a preservative. In another embodiment, a drug delivery system for a hydrophobic pharmaceutical compound can also be used. For example, the present invention may use a liposome and an emulsifier as a vehicle or carrier for administration. In a particular embodiment, an organic solvent such as N-decylpyrrolidone can be used. In still other embodiments, the compounds described herein can be administered using a sustained release system, such as a semipermeable matrix of a solid hydrophobic polymer containing a therapeutic agent. In addition, various sustained release materials can also be used in the present invention. In some embodiments, the sustained release capsule can release the compound over a period of hours, 67 201247612 or even over 24 hours. Additional protein stabilizers may be added depending on the chemical nature and biostability of the therapeutic agent. In a particular embodiment, the agents of the present invention may include one or more of an antioxidant, a metal chelating agent, a thiol containing compound, and/or other commonly used stabilizers. Examples of such stabilizers include, but are not limited to, (a) from about 0.5% to about 2% w/v glycerol, (b) from about 0.1% to about 1% w/v of methionine, ( c) from about 0.1% to about 2% w/v of monothioglycerol, (d) from about 1 mM to about 10 mM EDTA, (e) from about 0.01% to about 2% w/v of ascorbic acid, (f 0.003% to about 0.02% w/v of polysorbate 80 (p〇lysorbate 80), (g) 0.001. /. Up to about 0.05% w/v of polysorbate 20, (h) arginine, (1) heparin, (1) dextran sulfate, (k) cyclodextin, (1) pentose polysulfate and other heparinoids, (m) divalent cations such as magnesium ions and zinc ions; or (η) mixtures thereof. [Examples] Example 1: Demonstration of Preparation of Cyclohexenone Compounds Compound 1 (i.e., Antroq) was isolated from a solid-state fermenting mycelium of Antrodia camphorata by a conventional method (Lee, et al., Med, 73: 1412-1415, 2007). Based on previous research and experience, the effective dose of Compound 1 is 10-50 mg/kg body weight (Chang, et al.,

Comp/eweW d/iernai Med,2008) °除非特別註明,下述實驗 係以10-50 mg/kg體重之劑量進行試驗。 68 201247612 此外,示範化合物1可由4-羥基-2,3-二甲氧基-6-甲基 -2,5-環己稀明(4-hydroxy-2,3-dimethoxy-6-methyl-cyclo hexa-2,5-dienone)或其相似物所製備而得。 再者’其他具有Comp/eweW d/iernai Med, 2008) ° Unless otherwise specified, the following experiments were conducted at a dose of 10-50 mg/kg body weight. 68 201247612 In addition, the exemplary compound 1 can be 4-hydroxy-2,3-dimethoxy-6-methyl-2,5-cyclohexane (4-hydroxy-2,3-dimethoxy-6-methyl-cyclo Prepared from hexa-2,5-dienone) or its analogs. Again, the other has

結構之環己烯酮化 合物,亦可由牛樟芝分離而得,或以合成或半合成法從適 當起始物製備而得。熟知本技術領域者可在合成過程中使 用適當條件。 實施例2 ::局部性腎絲球硬化症(FSGS)之模型建立 本實驗係使用8週大BALB/c雌鼠進行試驗。FSGS小鼠 係以靜脈注射阿黴素(adriamycin)(0.1 mg/10 g體重)。在阿 黴素注射前六小時,係先餵食小鼠化合物1。在此,腹腔注 射生理食鹽水之BALB/c雌鼠係做為正控制組,而餵食载劑 (玉米油)之ASLN小鼠係做為控制組。於誘發疾病之7、14 或21天後犧牲老鼠,收集脾臟、腎皮質組織、及血液樣本, 並做適當保存以進行後續分析。所有的實驗均受到台灣國 防醫學院動物實驗管理委員會許可進行,且依據實驗動物 護理及使用NIH指南進行。 臨床及腎功能評估 於飼養3、7、14及21天時,收集飼養籠中尿液樣本, 並依據以前已知方法檢測樣本中之尿蛋白(Shui, et al., Ζ)ζ·α/ 2 1: 1 794-1 802, 2006)。此外,第 7、 14或21天犧牲小鼠,並收集小鼠血清樣本以測量血清中血 69 201247612 尿素氮(blood urea nitrogen, BUN)及血清肌酸酐(creatinine,The cyclohexenone compound of the structure may also be obtained by isolating from Antrodia camphorata or by synthetic or semi-synthetic methods from a suitable starting material. Those skilled in the art will be able to employ appropriate conditions in the synthesis process. Example 2: Model establishment of localized glomerulosclerosis (FSGS) This experiment was conducted using an 8-week-old BALB/c female. FSGS mice were given intravenous adriamycin (0.1 mg/10 g body weight). Six hours before the doxorubicin injection, the mouse compound 1 was fed first. Here, the BALB/c female mice in which the physiological saline was intraperitoneally injected were used as the positive control group, and the ASLN mice fed the vehicle (corn oil) were used as the control group. Mice were sacrificed after 7, 14 or 21 days of disease induction, and spleens, renal cortical tissue, and blood samples were collected and appropriately preserved for subsequent analysis. All experiments were approved by the Animal Experimental Management Committee of the Taiwan National Defense Medical College and were based on laboratory animal care and NIH guidelines. Clinical and renal function assessment During the 3, 7, 14 and 21 days of feeding, urine samples were collected from the cage and tested for urine protein (Shui, et al., Ζ)ζ·α/ according to previously known methods. 2 1: 1 794-1 802, 2006). In addition, mice were sacrificed on day 7, 14 or 21 and mouse serum samples were collected to measure blood in the serum. 69 201247612 Blood urea nitrogen (BUN) and serum creatinine (creatinine,

Cr)的含量。 腎臟病變評估 在此,係使用福馬林固定及石蠟包埋之腎臟切片,以 Shui et al.所述之方法,做腎臟病變及腎臟損傷程度之評估 (Shui,et al., 150: 216-222,2007)。腎臟病變及 腎臟損害分數評估,係依照Shui所述之方法《此外’取至 少50個腎臟組織切片之腎絲球,以評估内皮增生損害 (epithelial hyperplasia lesion, EPHL)及硬化。具有EPHL之 腎絲球數目,係根據先前所揭示之方法(例如,請參考Shui, et al., Nephrol Dial Transplant, 21: 1794-1802, 2006; Ka, et al·,iVepAro/ Ζ)ζ·α/ 21: 288-298, 2006),以總評估 腎絲球數目之百分比表示之β 此外,更進行組織免疫染色(immunohistochemistry, IHC)實驗。將以福馬林固定及石蠟包埋處理之腎臟切片, 與一級抗體、生物素標記第二抗體(Dako, Glostrup, Denmark)、以及抗生物素蛋白-生物素-過氧化酶複合物 (avidin-biotin-peroxidase complex)(Dako)反應;其中一級抗 體係辨識老鼠肌間線蛋白(Desmin)(Lab Vision, CA, USA)、CD3(pan-T cell; Serotec,NC,USA)、F4/80(單核白 血球/巨嗔細胞;Serotec)、IL-6 (R&D Systems, MN,USA)、 NF-κΒ p65(Cell Signaling Technology, MA,USA)、勝原蛋 白第 I、III、及 IV 型(Southern Biotech, AL,USA)、或 TGF-pl(Santa Cruz Biotechnology, CA, USA) °The content of Cr). Renal lesion assessment Here, the use of formalin-fixed and paraffin-embedded kidney sections was used to assess the extent of kidney disease and kidney damage as described by Shui et al. (Shui, et al., 150: 216-222). , 2007). Renal lesions and renal damage scores were evaluated according to the method described by Shui. In addition, at least 50 kidney skeins of kidney tissue sections were taken to evaluate epithelial hyperplasia lesion (EPHL) and sclerosis. The number of glomeruli with EPHL is based on previously disclosed methods (for example, see Shui, et al., Nephrol Dial Transplant, 21: 1794-1802, 2006; Ka, et al, iVepAro/ Ζ) ζ α/ 21: 288-298, 2006), expressed as a percentage of the total number of kidney spheroids. In addition, immunohistochemistry (IHC) experiments were performed. Renal sections treated with formalin-fixed and paraffin-embedded, with primary antibody, biotinylated secondary antibody (Dako, Glostrup, Denmark), and avidin-biotin-peroxidase complex (avidin-biotin) -peroxidase complex) (Dako) reaction; the primary anti-system recognizes mouse interstitial protein (Desmin) (Lab Vision, CA, USA), CD3 (pan-T cell; Serotec, NC, USA), F4/80 (single) Nuclear leukocytes/macellus cells; Serotec), IL-6 (R&D Systems, MN, USA), NF-κΒ p65 (Cell Signaling Technology, MA, USA), serotypes I, III, and IV (Southern) Biotech, AL, USA), or TGF-pl (Santa Cruz Biotechnology, CA, USA) °

實施例3 :評估ROS及NO 201247612 在此,係根據習知方法(Wu,et al.,Α^ρΛγο/ D/a/ Transplant, 2008, 23: 3082-3090, or Ka, et al., J. Am. Soc. Nephrol. 2007, 18:2473-2485)使用二氬乙錠 (dihydroethidium,DHE)染色檢測腎臟原位超氧化陰離子產 物。並透過螢光影像測量,以定量每單位腎臟切面總細胞 核之染色細胞核百分比。此外,以習知方法(Wu,et al., J Pineal Res, 2001, 30: 147-156, or Ka, et al., J. Am. Soc. Wep/iro/. 2007,18:2473-2485)測量血清及腎臟組織中之超 氧陰離子。血清、尿液、及腎臟組織中之ROS量量測,係 將樣品與Krebs-HEPES緩衝溶液反應,並以1.25mM亮光素 (lucigenin) (Sigma.:-Aldrich Chemical Co, MO)做為基質。螢 光計數則使用微孔盤發光檢測儀(Hidex Microplate Luminometer,Finland),以習知方法進行測量(Kretzler,et al.,. F/rchowijrc/z, 425: 181-193, 1994)。此外,超氧陰離 子係以每毫克乾燥組織之重量中,以每15分鐘之相對螢光 單位(RLU)表示,即,RLU/15分鐘/mg或RLU/15分鐘/m卜 血清中之NO含量,貝ij使用NO檢測套組(iNtRON Biotechnology, Seongnam,Korea),依據使用手冊,以重氮 化量(Griess方法)表示之。 實施例4::測量腎臟中細胞GPx活性 使用商業可購得之榖胱甘肽過氡化物酶檢測套組 (Glutathione Peroxidase Assay kit (Cayman,MI,USA))及其 使用手冊,測量腎臟組織中GPx活性。在此,酵素活性係以 相對於腎絲球均勻漿(glomerular homogenates)之蛋白質濃 度表示。 71 201247612 實施例5 : Nrf2及p47ph<)X之西方墨點分析 使用核蛋白萃取套組(Active Motif, Tokyo, Japan),依 其使用手冊萃取腎臟組織中之細胞質及細胞核蛋白。並使 用抗老鼠Nrf2、或p47ph°x(Santa Cruz)之兔子抗體,以西方 墨點分析測量腎臟組織中細胞質及/或細胞核之目標蛋白 質。在此,係分別使用組蛋白H3(Cell Signaling, CO, USA) 及β-肌動蛋白(Santa Cruz)抗體,測量細胞核及細胞質目標 蛋白之家管蛋白。 實施例6 :測量TGF-pi 在此,係使用商業購得之ELISA套組(R&D Systems), 並依其使用手冊檢測血清及腎臟組織中TGF-βΙ蛋白量。在 此,係使用I N HC1酸化樣品,並使用1.2 N NaOH/0.5 Μ HEPES中和酸化後之樣品,以檢測TGF-βΙ蛋白量。 實施例7 : B細胞缺陷小鼠之AcP-IgAN模型 B細胞缺陷小鼠(B6.129S2-Igh-6tmlCgn/J)係由中央研 究院分子生物研究所所提供(JohnT. Kung教授),且在台灣 台北國家衛生研究院動物中心飼養。依照先前實驗(Chao, et al.,尤ί办叮/«Λ 70:283-297; 2006),透過每日注射經純化之 IgA抗碟酿膽驗抗體及肺炎球菌C-多糖(pneumococcal C-p〇lysaccharide,PnC),以誘導小鼠之AcP-IgAN。所有的 實驗均受到台灣國防醫學院動物實驗管理委員會許可進 行,且依據實驗動物護理及使用NIH指南進行。 臨床及病理評估 72 201247612 每週測量小鼠的體重。每週收集飼養籠中尿液樣本, 且依據以前已知方法檢測樣本中之尿蛋白(Chao, et al., /πί. 70:283-297; 2006)。於第 3及 28天收集血清樣 品,以測量金清中血尿素氮(BUN)及血清肌酸酐(Cr)的含 量0 至於腎臟的組織病理’係使用10%福馬林緩衝液固定 組織,並以石蠟包埋樣品;而後,使用蘇木素伊紅染色法 (H&E)處理並將切片染色(4 μιη)。於光學顯微鏡下,以400X 放大備率,計算50個隨機腎絲球樣品中具有增生、新月型 (crescent formation)、硬化、或腎發炎(peri-glomerular inflammation)之腎絲球比例。 實施例8::免疫螢光染色(IF)、免疫组織化學技術(IHC)、及 細胞瑪亡福測 依照先前方法處理冷凍腎臟組織,並與異硫氰酸熒光 素(FITC)·共軛羊抗老鼠IgA或C3抗體反應(Cappel,NC),以 進行免疫螢光染色。染色強度計分則使用先前已知方式進 行(Ch.ao, et al.,尺/«Λ 70:283-297 (2006))。 福馬林固定及石蠟包埋處理之腎臟切片或冷凍切片, 與抗IL-6 (R&D Systems,MN,USA)、MCP-1 (Santa Cruz, CA)、F4/80(單核白血球/巨噬細胞;Serotec,NC)、膠原蛋白 第 I IV型(Southern Biotech,AL)、TGF_pi(Santa Cruz)、鱗 酸化 NF-k:B p65(Cell Signaling, MA)、CD3(pan-T cell ; Serotec) . CD4(輔助性T細胞;BioLegend,CA)、CD8(細胞 毒T細胞)、CDllb(巨噬細胞/中性粒細胞)、或CDllc (樹突 73 201247612 細胞)(BD Biosciences,CA)抗體反應,以進行IHC試驗。接 著,將切片與FITC-共耗、AlexaFluor 488-共輛(Invitrogen, CA)或辣根過氧化物酶(HRP)共軛二級抗體(DAKO, Denmark)反應。蘇木素或4·,6-二脒基-2·笨基吲哚 (4’,6-diamidino-2-phenylindole) (DAK0)則用以計算染色的 細胞核。 以末端去氧核苷酸轉移酶-相關dUTP缺口末端標記法 (TUNEL)進行細胞凋亡試驗。將福馬林固定及石蠟包埋處 理之組織切片,以ApopTag加氧酶原位細胞调亡摘測套組 (ApopTag plus peroxidase in situ apoptosis detection kit) (Chemicon,CA),依照使用手冊染色。測量腎臟皮質區域(包 括腎絲球、上腎絲球(peri-glomerular)區域),並以細胞/腎 絲球切面方式表示計分。 實施例9 :流式細胞儀 以Tris緩衝氣化銨處理老鼠的單球核(Splenocyte),已 去除紅血球,清洗後以含有10%胎牛血清、HEPES緩衝液、 L-麩酿胺酸及盤尼西林/鏈黴素(均購自Invitrogen)之RPMI 1640再懸浮。以T或B細胞活化表面標記將細胞染色。FITC· 共軛抗小鼠CD3、CD4、CD8、或CD19(B細胞)抗體與藻紅 蛋白(phycoerythrin,PE)共軛抗小鼠CD69抗體(均購自BD Biosciences),係以 FACSCalibur (BD Biosciences)進行分析。 實施例10 : T細胞增生分析 依照前述方法處理老鼠的單球核,而後於96孔平底微 孔盤,於每孔中培養細胞(200 μΐ/孔中含有5 X 105細胞),重 複三次,其中微孔盤中係先塗佈有0.25 pg/ml之抗小鼠CD3 74 201247612 抗體(BD Biosciences)於4°C下靜置隔夜。於48小時,以1 μΟΜ 之3Η-甲基胸腺啦咬脱氧核普(Amersham Pharmacia Biotech, NJ)處理培養細胞,於1 6小時後收集,並使用TopCount (Packard,PerkinElmer,MA)測量細胞所吸收之3H-甲基胸腺 ,咬脱氧核普。 實施例11 : IL-Ιβ、IL-6、IL-18及MCP-1之酵素連結免疫吸 附分析法(ELISA) 根據使用手冊,以商業購得之ELISA套組測量 IL-1 P(eBi.osceience, CA) ' IL-6(eBiosceience) ' IL-18(MBL, Japan)及MCP-l(eBiosceience)。 使用細胞核萃取套組(Active Motif, Japan)萃取核蛋 白。使用 Trans-AM ELIS A 套組 Trans-AM ELISA,並依其使 用手冊,測量腎組織核蛋白萃取物中之磷酸化NF-κΒ p65及 腎細胞核因數-E2相關因數2 (Nrf2)。使用RIPA緩衝液(Cell signaling)萃取腎細胞蛋白。 細胞榖胱甘肽過氧化酶(GPx) (Cayman,MI)及細胞第 一髮血色素氧化酵素(HO-1) (Enzo Life Sciences, NY)’ 則 依據商業購得知ELISA套組及其使用手冊進行測量。此兩者 係以相對於細胞裂解液蛋白質濃度表示。 於所有的ELISA試驗中,使用ELISA讀取儀(Bio-Tek, ΜΑ)測量於450 nm之吸光值。 實施例12 :即時PCR分析Example 3: Evaluation of ROS and NO 201247612 Here, according to conventional methods (Wu, et al., Α^ρΛγο/ D/a/Transplant, 2008, 23: 3082-3090, or Ka, et al., J Am. Soc. Nephrol. 2007, 18: 2473-2485) The in situ superoxide anion product of the kidney was detected by dihydroethidium (DHE) staining. Fluorescence images were measured to quantify the percentage of stained nuclei per cell kidney total cell nuclei. In addition, by conventional methods (Wu, et al., J Pineal Res, 2001, 30: 147-156, or Ka, et al., J. Am. Soc. Wep/iro/. 2007, 18: 2473-2485 ) Measurement of superoxide anion in serum and kidney tissue. The amount of ROS in serum, urine, and kidney tissues was measured by reacting the sample with a Krebs-HEPES buffer solution and using 1.25 mM lucigenin (Sigma.: - Aldrich Chemical Co, MO) as a substrate. The fluorescence count was measured by a known method using a Hidex Microplate Luminometer (Finland) (Kretzler, et al., F/rchowijrc/z, 425: 181-193, 1994). In addition, superoxide anion is expressed in terms of relative fluorescence units (RLU) per 15 minutes in weight per milligram of dry tissue, ie, RLU/15 minutes/mg or RLU/15 minutes/m. Bayer ij uses the NO test kit (iNtRON Biotechnology, Seongnam, Korea), which is expressed in terms of diazotization (Griess method) according to the manual. Example 4: Measurement of cellular GPx activity in kidneys A commercially available glutathione Peroxidase Assay kit (Cayman, MI, USA) and its manual were used to measure renal tissue. GPx activity. Here, the enzyme activity is expressed as a protein concentration relative to glomerular homogenates. 71 201247612 Example 5: Western blot analysis of Nrf2 and p47ph<) X The cytoplasm and nuclear protein in kidney tissue were extracted using a nuclear protein extraction kit (Active Motif, Tokyo, Japan) according to its manual. The target protein of the cytoplasm and/or nucleus in the kidney tissue was measured by western blot analysis using a rabbit antibody against mouse Nrf2 or p47ph°x (Santa Cruz). Here, the histone protein of the nuclear and cytoplasmic target proteins was measured using histone H3 (Cell Signaling, CO, USA) and β-actin (Santa Cruz) antibodies, respectively. Example 6: Measurement of TGF-pi Here, a commercially available ELISA kit (R&D Systems) was used, and the amount of TGF-βΙ protein in serum and kidney tissues was measured according to its manual. Here, the sample was acidified using I N HC1, and the acidified sample was neutralized using 1.2 N NaOH / 0.5 Μ HEPES to detect the amount of TGF-β prion protein. Example 7: AcP-IgAN model of B cell deficient mice B cell deficient mice (B6.129S2-Igh-6tmlCgn/J) were provided by the Institute of Molecular Biology, Academia Sinica (Prof. John T. Kung), and Animal Center of Taipei National Institute of Health, Taiwan. According to previous experiments (Chao, et al., 尤 叮 Λ / «Λ 70:283-297; 2006), through daily injection of purified IgA anti-battering test antibody and pneumococcal C-polysaccharide (pneumococcal Cp〇) Lysaccharide, PnC), to induce AcP-IgAN in mice. All experiments were approved by the Animal Experimental Management Committee of the Taiwan National Defense Medical College and were based on laboratory animal care and NIH guidelines. Clinical and pathological assessment 72 201247612 The body weight of mice was measured weekly. Urine samples from cages were collected weekly and urine samples were tested in samples according to previously known methods (Chao, et al., /πί. 70:283-297; 2006). Serum samples were collected on days 3 and 28 to measure the content of blood urea nitrogen (BUN) and serum creatinine (Cr) in Jinqing. 0 As for the histopathology of the kidney, the tissue was fixed with 10% formalin buffer and wrapped in paraffin. The sample was buried; then, hematoxylin and eosin staining (H&E) was used and the sections were stained (4 μιη). The proportion of renal spheroids with hyperplasia, crescent formation, sclerosis, or peri-glomerular inflammation in 50 random kidney spheroid samples was calculated under a light microscope at 400X magnification. Example 8: Immunofluorescence staining (IF), immunohistochemistry (IHC), and cell death assay. Frozen kidney tissue was treated according to the previous method and conjugated with fluorescein isothiocyanate (FITC). Goat anti-mouse IgA or C3 antibody reaction (Cappel, NC) for immunofluorescence staining. The staining intensity score is performed using a previously known method (Ch. Ao, et al., Ruler / «Λ 70:283-297 (2006)). Renal sections or cryosections of formalin-fixed and paraffin-embedded treatments, with anti-IL-6 (R&D Systems, MN, USA), MCP-1 (Santa Cruz, CA), F4/80 (mononuclear leukocytes/giant) Phage; Serotec, NC), collagen type I IV (Southern Biotech, AL), TGF_pi (Santa Cruz), squamized NF-k: B p65 (Cell Signaling, MA), CD3 (pan-T cell; Serotec CD4 (helper T cells; BioLegend, CA), CD8 (cytotoxic T cells), CD11b (macrophage/neutrophils), or CDllc (dendritic 73 201247612 cells) (BD Biosciences, CA) antibody The reaction was carried out for the IHC test. Next, the sections were reacted with FITC-co-consumed, AlexaFluor 488-common (Invitrogen, CA) or horseradish peroxidase (HRP) conjugated secondary antibody (DAKO, Denmark). Hematoxylin or 4,6-diamidino-2-phenylindole (DAK0) was used to calculate the stained nuclei. Apoptosis assays were performed with terminal deoxynucleotidyl transferase-related dUTP nick end labeling (TUNEL). The tissue of the formalin-fixed and paraffin-embedded tissues was sectioned and stained according to the manual using the ApopTag plus peroxidase in situ apoptosis detection kit (Chemicon, CA). The renal cortical area (including the renal pelvis and the peri-glomerular area) was measured and scored in a cell/kidney spheroidal section. Example 9: Flow cytometry The single-core nuclei (Splenocyte) of mice were treated with Tris buffered ammonium hydride. The red blood cells were removed and washed to contain 10% fetal bovine serum, HEPES buffer, L-branched tyrosine and penicillin. Resuspension of RPMI 1640 / streptomycin (both purchased from Invitrogen). Cells were stained with T or B cell activated surface markers. FITC·conjugated anti-mouse CD3, CD4, CD8, or CD19 (B cell) antibody and phycoerythrin (PE) conjugated anti-mouse CD69 antibody (both purchased from BD Biosciences) with FACSCalibur (BD Biosciences) ) for analysis. Example 10: T cell proliferation assay The single core of the mouse was treated according to the method described above, and then the cells were cultured in each well (200 μΐ/well containing 5 X 105 cells) in a 96-well flat-bottom microplate, and repeated three times. The microporous disc was first coated with 0.25 pg/ml of anti-mouse CD3 74 201247612 antibody (BD Biosciences) and allowed to stand overnight at 4 °C. At 48 hours, the cultured cells were treated with 1 μΟΜ of 3Η-methylthymidine deoxynucleotide (Amersham Pharmacia Biotech, NJ), collected after 16 hours, and measured for cell uptake using TopCount (Packard, PerkinElmer, MA). 3H-methyl thymus, bite deoxynucleotide. Example 11: Enzyme-linked immunosorbent assay (ELISA) for IL-Ιβ, IL-6, IL-18, and MCP-1 IL-1 P (eBi.osceience) was measured using a commercially available ELISA kit according to the instruction manual. , CA) 'IL-6 (eBiosceience) ' IL-18 (MBL, Japan) and MCP-l (eBiosceience). Nucleoproteins were extracted using a nuclear extraction kit (Active Motif, Japan). The Trans-AM ELIS A kit Trans-AM ELISA was used and the phosphorylated NF-κΒ p65 and renal cell nuclear factor-E2 correlation factor 2 (Nrf2) in renal tissue nuclear protein extracts were measured according to their manual. Renal cell proteins were extracted using RIPA buffer (Cell signaling). Cellular glutathione peroxidase (GPx) (Cayman, MI) and cell first hemoglobin oxidase (HO-1) (Enzo Life Sciences, NY)' were based on commercially available ELISA kits and their manuals. Make measurements. Both are expressed relative to the cell lysate protein concentration. Absorbance at 450 nm was measured using an ELISA reader (Bio-Tek, ΜΑ) in all ELISA assays. Example 12: Real-time PCR analysis

總腎皮質RNA係使用TriZOL試劑(Invitrogen),由腎皮 質上萃取而得。第一股cDNA合成,則使用1.5 pg之總RNA 75 201247612 進行第一次反轉錄反應。其中,反應混合物包括0.9 pg之12 至18單元寡dT引子,1.0 mM之去氧核三磷酸(dNTP)、1 pg 之第一股緩衝液、0.4 mM之二硫甦糖醇、80 U之去RNase 複合核醣核酸酶抑制劑、及300 U之超轉錄II RNase Η (Invitrogen)。即時 PCR係以 ABI Prism 7700序列债測儀(ΑΒΙ Prism 7700 Sequence Detection System, Applied Biosystems, CA)進行。所有的探針及引子,均為Assays-on-Demand Gene expression之產品(Applied Biosystems)。即時PCR反應係使 用 10 μΐ之cDNA、12.5 μΐ之TaqMan Universal PCR Master Mix(Applied Biosystems)、及1.25 μΐ之特殊探針/引子之混 合物,其總體積為25 μΐ。加熱循環設定係為:50°C進行2 分鐘,95°C進行10分鐘,進行DNA分開40個循環(95°C進行 15秒),而後進行黏合/延伸反應(60°C進行1分鐘)》 實施例13 :西方墨點分析 以10% SDS-PAGE膠體分離每一蛋白樣品。將膠體電 轉印製聚二氟乙烯硝酸纖維素膜(Amersham Int.,UK);以封 閉緩衝液(Tris-緩衝生理食鹽水,其含有5%脫脂牛奶)固定1 小時;並於4°C下與兔子之抗nacht區-(nacht domain-)、富含 亮氨酸重複-(leucine-rich repeat-)、及含pyrin區蛋白3 (pyrin domain (PYD)-containing protein 3, NLRP3)、调亡因 子-1 (caspase-1)或β-肌動蛋白抗體(均購自Santa Cruz)反應 隔夜。清洗後,將膜與HRP-共軛羊抗老鼠(DAKO)抗體,在 室溫下反應1小時。將化學冷光試劑(chemiluminescent reagent plus, PerkinElmer Life Sciences,ΜΑ)與結合有抗體 的膜反應,並以x光照相進行測量。 76 201247612 實施例14 ::數據分析 在此,係使用學生1;測試(Student’s i test)進行兩組間數 據分析比較,結果係以平均值±SEM表示。其中,多組間的 差異則以單項變方分析(One-way Analysis of Variance, ANOVA),並使用Tukey’s法在進行事後檢定分析。在此, p<0.05表示具有統計顯著差異。 FSGS模型 於疾病控制組中,僅有經載劑處理之FSGS小鼠(FSGS 控制組),從第7天起尿蛋白量顯著增加,且直到實驗結束 之第21天仍觀察到持續增加的現象,如圖1A所示。然而, 經化合物1處理之FSGS小鼠(FSGS+Antroq小鼠),此現象係 明顯受到抑制,且尿蛋白之增加量與正常小鼠相似。此外, 相較於控制組FSGS小鼠,從第14天至第21天,可觀察到血 清中企尿素氮(blood urea nitrogen, BUN)(圖1B)及血清肌酸 酐(creatinine,Cr)(圖1C)含量顯著且持續增加;但 FSGS+Antroq小鼠(即,化合物1),其腎功能有顯著提升。 FSGS+Antroq小鼠在第7天時,其BUN或Cr量與正控制組小 鼠(FSGS控制組)無顯著差異。 此外,更於不同時間點對腎躂切片做組織病理學檢驗 (如圖2A所示)。相較於正常控制組小鼠,在FSGS控制組之 小鼠中,第7天僅觀察到些微腎絲球之胞外間質擴散及群 聚,且從第14天起至第21天可逐漸看到明顯的組織損傷。 值得注意的是,從第14天起至第·21天,可觀察到包含EPHL 及腎絲球旁單核浸潤現象比例顯著且持續提升,此代表著 77 201247612 進行式病理狀態。相反的,於FSGS+Antroq小鼠中,這些進 行式腎衰竭現象係大幅減少。此外,足細胞(podocyte)損傷 及消失係為一種可判斷FSGS是否發展的病理標記。為了檢 驗Antroq處理之FSGS足細胞之表現變化,在此係使用組織 免疫染色法檢驗肌間線蛋白(Desmin)之表現,此係為辨識足 細胞之標記。如圖2B所示,在第14天及第21天,相較於FSGS 控制組小鼠,FSGS+Antroq小鼠之肌間線蛋白表現可顯著大 幅抑制,即便正常控制組小鼠在21天仍可觀察到些微肌間 線蛋白表現的現象。 這些結果顯示,示例化合物1可抑制尿蛋白增加、提升 腎臟功能、改善包括内皮增生衰竭(EPHL)之腎衰竭、及改 善腎絲球損傷之功效。 於血清及尿液中系统性抑制氧化壓力之效果 相較於正常控制組小鼠,於FSGS控制組小鼠(FSGS + 載劑)中,血清中超氧化陰離子量於第7及14天時係明顯增 加,而於第21天時稍微減少,但仍稍高於正常控制組小鼠。 然而,於第7天至第21天當投予示例化合物1 (Antroq),可 有效調整超氧化陰離子之產生,且超氧化陰離子量係與正 常控制組小鼠差不多,如圖3A所示。此外,FSGS控制組小 鼠中,血清中NO量於第7天時明顯上升,且在第14天及第 21天時均觀察到高濃度的情形。當投予FSGS小鼠示例化合 物l(FSGS+Antroq),則可顯著抑制高的血清NO量,如圖3B 所示。相較於正常組小鼠於FSGS控制組小鼠中,在第7天 可觀察到尿液中超氧化陰離子量顯著增加,且維持到地21 78 201247612 天。反之,在FSGS+Antroq小鼠中,當投予示例化合物1可 明顯降低超氧化陰離子量,如圖3C所示。雖然FSGS控制組 小鼠中尿液的NO量於第14天至第21天有明顯增加,但當投 予示例化合物1可減緩FSGS+Antroq小鼠中此現象發生,如 圖3D所示。而在第7天時,並未觀察到正常控制組、FSGS+ 載劑、及FSGS+Antroq小鼠尿液中NO量有顯著差別。 於腎臟组織中局部抑制ROS產生 如圖3E所示,相較於正常控制組小鼠,FSGS控制組小 鼠之腎臟中超氧化陰離子量在第14天時可觀察到明顯增 加,且持續增加至第21天。然而,於第14天至第21天時, 可觀察到投予Antroq可有效調整超氧化陰離子之產生,且 超氧化陰離子量係與正常控制組小鼠差不多。 為了更加了解ROS於腎贜中產生的位置,使用二氫乙 錠(DHE)試驗檢測腎臟組織原位ROS產物。如圖3F所示,於 FSGS控制組小鼠之腎臟組織中,從第14天至第21天,DHE 螢光量有增加,特別是在腎絲球及腎小管部分有顯著增 加,表示相較於正常組小鼠有原位ROS的產生。反之,在 第14天至第21天,FSGS+Antroq小鼠僅觀察到非常少量的 DHE螢光量。The total renal cortical RNA was obtained by extracting from the kidney cortex using TriZOL reagent (Invitrogen). For the first cDNA synthesis, the first reverse transcription reaction was performed using 1.5 pg of total RNA 75 201247612. Wherein, the reaction mixture comprises 0.9 pg of 12 to 18 unit oligo dT primer, 1.0 mM deoxyribonucleoside (dNTP), 1 pg of first buffer, 0.4 mM dithiothreitol, 80 U RNase complex ribonuclease inhibitor, and 300 U of super-transcription II RNase Η (Invitrogen). The real-time PCR was performed on an ABI Prism 7700 Sequence Debt Detector (ΑΒΙ Prism 7700 Sequence Detection System, Applied Biosystems, CA). All probes and primers are products of Assays-on-Demand Gene expression (Applied Biosystems). The real-time PCR reaction used a mixture of 10 μΐ of cDNA, 12.5 μM of TaqMan Universal PCR Master Mix (Applied Biosystems), and a special probe/primer of 1.25 μM in a total volume of 25 μM. The heating cycle was set at 50 ° C for 2 minutes, 95 ° C for 10 minutes, DNA separation for 40 cycles (95 ° C for 15 seconds), and then adhesion / extension reaction (60 ° C for 1 minute) Example 13: Western blot analysis Each protein sample was separated by 10% SDS-PAGE colloid. The colloidal electrotransfer was made into a polyvinylidene fluoride nitrocellulose membrane (Amersham Int., UK); fixed in blocking buffer (Tris-buffered physiological saline containing 5% skim milk) for 1 hour; and at 4 ° C And rabbit anti-nacht region-(nacht domain-), leucine-rich repeat-, and pyrin domain (PYD)-containing protein 3 (NLRP3), apoptosis Factor-1 (caspase-1) or β-actin antibodies (both purchased from Santa Cruz) were reacted overnight. After washing, the membrane was reacted with HRP-conjugated goat anti-mouse (DAKO) antibody for 1 hour at room temperature. A chemical luminescence reagent (chemiluminescent reagent plus, PerkinElmer Life Sciences, ΜΑ) was reacted with an antibody-bound membrane and measured by x-ray photography. 76 201247612 Example 14 :: Data Analysis Here, Student 1 s i test was used to perform data analysis comparison between the two groups, and the results were expressed as mean ± SEM. Among them, the differences between groups were analyzed by One-way Analysis of Variance (ANOVA) and Tukey's method was used for post-mortem analysis. Here, p < 0.05 indicates a statistically significant difference. The FSGS model in the disease control group, only vehicle-treated FSGS mice (FSGS control group), the amount of urinary protein increased significantly from day 7, and continued to increase until the 21st day of the end of the experiment. , as shown in Figure 1A. However, in the compound 1 treated FSGS mice (FSGS + Antroq mice), this phenomenon was significantly inhibited, and the increase in urine protein was similar to that in normal mice. In addition, compared with the control group FSGS mice, blood urea nitrogen (BUN) (Fig. 1B) and serum creatinine (Cr) were observed from day 14 to day 21 (Fig. 1C) The content was significantly and continuously increased; however, FSGS+Antroq mice (ie, Compound 1) had a significant increase in renal function. On day 7 of FSGS+Antroq mice, the amount of BUN or Cr was not significantly different from that of the control group (FSGS control group). In addition, histopathological examination of the renal pelvis section was performed at different time points (as shown in Fig. 2A). Compared with the normal control group of mice, in the FSGS control group, only the extracellular interstitial proliferation and aggregation of micro-renal spheroids were observed on the 7th day, and gradually increased from the 14th day to the 21st day. See obvious tissue damage. It is worth noting that from the 14th day to the 21st day, a significant and continuous increase in the proportion of mononuclear infiltration including EPHL and renal spheroids was observed, which represents the pathological state of 77 201247612. In contrast, in FSGS+Antroq mice, these progressive renal failures were significantly reduced. In addition, the podocyte injury and disappearance is a pathological marker that can determine whether FSGS is developing. In order to examine the changes in the expression of Antroq-treated FSGS podocytes, the expression of interstitial protein (Desmin) was examined using tissue immunostaining, which is a marker for identifying podocytes. As shown in Fig. 2B, on day 14 and day 21, the intermuscular protein expression of FSGS+Antroq mice was significantly inhibited significantly compared to the FSGS control group, even in the normal control group. Some phenomena of micro-muscle protein expression can be observed. These results show that the exemplified compound 1 inhibits an increase in urinary protein, enhances renal function, improves renal failure including endothelial proliferative failure (EPHL), and improves renal pelvic damage. The effect of systemic inhibition of oxidative stress in serum and urine compared with the normal control group of mice, in the FSGS control group (FSGS + vehicle), the amount of superoxide anion in serum was evident on days 7 and 14. Increased, but decreased slightly on day 21, but still slightly higher than the normal control group of mice. However, when Example Compound 1 (Antroq) was administered from Day 7 to Day 21, the production of superoxide anion was effectively adjusted, and the amount of superoxide anion was similar to that of the normal control group, as shown in Fig. 3A. In addition, in the FSGS control group, the amount of NO in the serum increased significantly on the 7th day, and a high concentration was observed on the 14th day and the 21st day. When FSGS mouse exemplified compound 1 (FSGS + Antroq) was administered, a high serum NO amount was significantly inhibited as shown in Fig. 3B. A significant increase in the amount of superoxide anion in the urine was observed on day 7 compared to the normal group of mice in the FSGS control group and maintained to the ground 21 78 201247612 days. On the contrary, in the FSGS + Antroq mice, when the exemplified compound 1 was administered, the amount of superoxide anion was remarkably lowered as shown in Fig. 3C. Although the amount of NO in the urine of the FSGS-controlled group was significantly increased from day 14 to day 21, administration of the exemplified compound 1 attenuated this phenomenon in FSGS+Antroq mice, as shown in Fig. 3D. On day 7, no significant difference in the amount of NO in the urine of the normal control group, FSGS+ carrier, and FSGS+Antroq mice was observed. Local inhibition of ROS production in kidney tissue As shown in Figure 3E, the amount of superoxide anion in the kidneys of the FSGS control group was observed to increase significantly on day 14 compared to the normal control group, and continued to increase to Day 21 However, from day 14 to day 21, it was observed that administration of Antroq effectively regulated the production of superoxide anion, and the amount of superoxide anion was similar to that of the normal control group. To better understand the location of ROS in the renal pelvis, the in situ ROS product of kidney tissue was measured using the dihydrogen ingot (DHE) assay. As shown in Fig. 3F, in the kidney tissues of the mice in the FSGS control group, the amount of DHE fluorescence increased from day 14 to day 21, especially in the renal glomeruli and renal tubules, indicating that compared with Normal group mice had in situ ROS production. On the other hand, on day 14 to day 21, only a very small amount of DHE fluorescence was observed in FSGS+Antroq mice.

Nrf-2相關抗氧化訊息傳遞路徑 透過測量腎臟中NAD(P)H氧化酶單元p47phux之蛋白表 現量、Nrf2轉移至細胞核(活化)、及GPx活性,以了解Antroq 於抗氧化訊息傳導路徑上之功效。 79 201247612 氣中,從第14天至第The Nrf-2-associated antioxidant signaling pathway is used to measure the expression of NAD(P)H oxidase unit p47phux in the kidney, Nrf2 transfer to the nucleus (activation), and GPx activity to understand Antroq's pathway in the antioxidant signaling pathway. efficacy. 79 201247612 In the air, from the 14th day to the first

如圖4A及4C所示,FSGS控制組/丨、胃 21天’可觀察到NAD(P)H氧化酶 明顯增加,而投予Antroq可抑制 於FSGS正常組小鼠或正常小鼠, 小鼠之财2轉移至細胞核情形明顯増加,且㈣維持到第 21天,如圖4B及4D所示。 此外’如圖4E所示’相較於正常控制組小鼠,^仍控 制組小鼠之GPx活性有顯著下降,其中GPx係為Nrf2之下游 II相酵素之一。然而,相較於FSGS控制組小鼠,FSGS+Antr〇q 小鼠在第7天時腎臟中GPx活性有恢復,且持續到第21天小 鼠被犧牲仍觀察到相同情形。 此結果顯示,示例化合物1可抑制ROS/NO產生,但增 加Nrf2活化及GPx活性。 T細胞及巨噬細胞浸潤 巨噬細胞及淋巴細胞之組織間質聚集(interstitial recruitment)係為評估FSGS發炎及纖維化(Profibrotic)因素 之一。在此,係評估示例化合物1對腎臟組織T細胞及/或單 核白血球/巨噬細胞浸潤之影響。於FSGS控制組小鼠中,在 第14天及第21天時,可明顯發現腎臟組織間之腎絲球區域 有明顯T細胞(CD3+)及單核白血球/巨噬細胞(F4/80+)浸潤現 象’但FSGS+Antroq小鼠卻與正常控制組小鼠相似,未觀察 到明顯的浸潤現象,如圓5A及5B所示。 IL-6表現 201247612 在此,係測量腎臟中IL-6產生。如圖6A之IHC結果所 示,在第7天時即可觀察到IL-6蛋白質表現亮明顯增加,且 直到小鼠犧牲之第21天仍持續上升。然而,相較於FSGS控 制組小鼠,FSGS+Antroq小鼠之腎臟中IL-6蛋白質表現量則 顯著降低> 抑制NF-κΒ活性 在此,係測量投予示例化合物1對腎臟組織NF-κΒ活性 之影響。如圖6B所示,FSGS控制組小鼠之核NF-κΒ p65表 現量在第14天及第21天顯著增加,而FSGS+Antroq小鼠之 NF-κΒ活性則顯著受到抑制。與IHC實驗結果相符,腎贜組 織核蛋白萃取物之ELI S A試驗亦顯示,相較於正常控制組小 鼠,FSGS控制組小鼠在第14天時可觀察到NF-κΒ p65核蛋 白表現量增加,且在第21天時仍顯著增加。然而,當投予 化合物1 (Antroq)第14天時,FSGS+Antroq小鼠之此現象係 稍稍受到抑制,而於第21天時明顯受到抑制。此外,於第7 天時,可觀察到FSGS控制組或FSGS+Antroq小鼠有明顯差 別,如圖6C所示。 因此,當投予本發明之環己烯酮化合物,可抑制腎臟 中NF-κΒ活化,而有效抑制發炎反應。 此外,亦觀察FSGS模型中,觀察纖維化相關之蛋白質 表現量,即膠原蛋白(collagen)I、III及IV,以了解投予環己 烯酮化合物對腎絲球纖維化之效果。如圖7A-7C所示,相較 於正常控制組小鼠,FSGS控制組小鼠於從第14天至第21天 可觀察到腎臟膠原蛋白I及IV明顯表現,第21天可觀察膠原 81 201247612 蛋白III ;而投予化合物1則可明顯抑制這類蛋白質的產生, 在FSGS+Antroq小鼠中之表現量與正常控制組小鼠表現量 無顯著差異。TGF-βΙ係為腎纖維化及新月型生成之一基礎 生長因子及生長激素》ELIS A結果顯示,相較於正常控制組 小鼠,在FSGS小鼠中,第14天時可觀察到血清中(圖8A)及 腎臟組織中(圖8B)TGF-pl蛋白量增加,而在第21天顯著提 升。然而,投予示例化合物1時,無論在血清或腎臟中,增 加之TGF-βΙ蛋白量可被抑制。在此,更以IHC試驗訂定腎 臟組織中TGF-βΙ蛋白表現量的變化。同樣的,IHC結果顯 示,FSGS+Antroq小鼠之腎臟中TGF-βΙ蛋白量明顯低於 FSGS控制組小鼠,如圖8C所示。因此,當使用本發明所提 供之環己烯酮化合物,可有效抑制TGF-βΙ蛋白表現,而可 應用於腎絲球硬化之治療上。As shown in Figures 4A and 4C, FSGS control group / sputum, stomach 21 days 'can observe a significant increase in NAD (P) H oxidase, while administration of Antroq can inhibit FSGS normal group mice or normal mice, mice The transfer of the 2nd to the nuclear condition was significantly increased, and (4) was maintained until the 21st day, as shown in Figures 4B and 4D. Further, as shown in Fig. 4E, the GPx activity of the mice in the control group was significantly decreased as compared with the mice in the normal control group, and the GPx system was one of the downstream phase II enzymes of Nrf2. However, compared with the FSGS control group, GPGS activity was restored in the FSGS+Antr〇q mice on day 7, and the same situation was observed until the 21st day when the mice were sacrificed. This result shows that the exemplified compound 1 can inhibit ROS/NO production, but increases Nrf2 activation and GPx activity. T cell and macrophage infiltration The interstitial recruitment of macrophages and lymphocytes is one of the factors that evaluate FSGS inflammation and fifi (Profibrotic). Here, the effect of the exemplified compound 1 on renal tissue T cells and/or mononuclear leukocyte/macrophage infiltration was evaluated. In the FSGS control group, on day 14 and day 21, significant T cells (CD3+) and mononuclear leukocytes/macrophages (F4/80+) were found in the renal spheroidal region between kidney tissues. Infiltration phenomenon, but FSGS+Antroq mice were similar to the normal control group, and no significant infiltration was observed, as shown by circles 5A and 5B. IL-6 Performance 201247612 Here, IL-6 production in the kidney is measured. As shown by the IHC results in Figure 6A, a significant increase in IL-6 protein expression was observed on day 7 and continued to rise until the 21st day of sacrifice. However, the expression of IL-6 protein in the kidney of FSGS+Antroq mice was significantly lower than that of the FSGS control group mice. Inhibition of NF-κΒ activity Here, the administration of exemplary compound 1 to kidney tissue NF- was measured. The effect of κΒ activity. As shown in Fig. 6B, the nuclear NF-κΒ p65 expression of the FSGS-controlled mice was significantly increased on the 14th day and the 21st day, while the NF-κΒ activity of the FSGS+Antroq mice was significantly inhibited. Consistent with the results of the IHC experiment, the ELI SA test of the nucleoprotein extract of renal pelvis also showed that NF-κΒ p65 nucleoprotein expression was observed on the 14th day in the FSGS control group compared with the normal control group. Increased and still increased significantly on day 21. However, this phenomenon of FSGS + Antroq mice was slightly inhibited when the compound 1 (Antroq) was administered on the 14th day, and was significantly inhibited on the 21st day. In addition, on day 7, it was observed that there was a significant difference between the FSGS control group or the FSGS+Antroq mice, as shown in Fig. 6C. Therefore, when the cyclohexenone compound of the present invention is administered, the activation of NF-κΒ in the kidney can be inhibited, and the inflammatory reaction can be effectively inhibited. In addition, the FSGS model was also observed to observe the protein expression of fibrosis, namely collagen I, III and IV, in order to understand the effect of administration of the cyclohexenone compound on renal fibrosis. As shown in Figures 7A-7C, kidney collagen I and IV were observed from day 14 to day 21 in the FSGS control group compared to the normal control group. Collagen 81 was observed on day 21. 201247612 Protein III; while administration of Compound 1 significantly inhibited the production of such proteins, and the amount of expression in FSGS+Antroq mice was not significantly different from that in the normal control group. TGF-β Ι is one of the basic growth factors and growth hormones of renal fibrosis and crescentic formation. ELIS A results show that serum can be observed on day 14 in FSGS mice compared with normal control mice. The amount of TGF-pl protein was increased in the middle (Fig. 8A) and in the kidney tissues (Fig. 8B), and was significantly increased on the 21st day. However, when the exemplified compound 1 was administered, the amount of the increased TGF-β prion protein was inhibited regardless of the serum or the kidney. Here, the change in the amount of TGF-βΙ protein expression in the renal tissues was determined by the IHC test. Similarly, IHC results showed that the amount of TGF-βΙ protein in the kidneys of FSGS+Antroq mice was significantly lower than that in the FSGS control group, as shown in Figure 8C. Therefore, when the cyclohexenone compound provided by the present invention is used, the expression of TGF-βΙ protein can be effectively inhibited, and it can be applied to the treatment of glomerular sclerosis.

IgAN模型 投予載劑之AcP-IgAN小鼠(AcP-IgAN控制組小鼠)係 作為疾病控制組,其在誘發疾病第7天有尿蛋白量增加,且 直到實驗結束之第28天也持續上升,如圓9所示》然而,以 化合物1處理之AcP-IgAN小鼠(AcP-IgAN+Antroq小鼠),此 情形係被大幅抑制,雖然相較於正常控制組仍有些微尿蛋 白產生。此外,AcP-IgAN控制組小鼠在第28天之BUN血清 量(圖9B)及血清肌酸酐量(圖9C)有顯著增加;雖然 AcP-IgAN+Antroq小鼠與AcP-IgAN控制組、正常組小鼠在 第3天時沒有顯著的BUN及Cr血清量差別,但 AcP-IgAN+Antroq小鼠卻展現較佳之腎功能。 82 201247612 每週測量小鼠的鱧重e AcP-IgAN+Antroq小鼠與 AcP-IgAN控制組小鼠之生長,與正常控制組小鼠並無差 異。此外,每一組的所有小鼠均展現正常活性,而無落髮 或食慾變化的情形。 腎臟病理試驗 如圖9A-9E所示,在第28天時,AcP-IgAN控制組小鼠 有與噬中性白血球浸潤、局部但典型的新月型、及/或腎絲 球節段性硬化等相關之浸潤增加現象,且有顯著的腎單核 白血球浸潤及散佈之腎小管萎縮(與蛋白脫落有關)現象產 生,此結果顯示相較於先前研究之腎絲球組織病理報告 (Lai, K.N. Nephron. 92:263-270 (2002) ; Lai, et al., Nephron. 69:1-8 (1995); Chen, et al., J. Clin. Lab. Analysis. 6:35-39 (1992); Kashem,et al”. /«ί· 45: 868-875 (1994))、及 腎間質組織病理研究(Falk,et al.,沿咖叮/«ί. 47:177-185 (1995); van Es, et al., Kidney Int. 73:1426-1433 (2008); Torres, et al., Kidney Int. 73:327-333 (2008); Walsh, et al., Clin J Am Soc. Nephrol. 5:425-430 (2010); Fujinaka, et al., J. #ep;7rc)/· 20:357-363 (2007)),有腎病增加且惡化的狀態》 除了腎絲球免疫沉澱所導致之腎損傷外,AcP-IgAN+Antroq 小鼠之其他所有腎臟損傷基本上均受到抑制。於第3天時, 控制組AcP-IgAN小鼠則開始有局部性腎病產生,但以化合 物1處理之AcP-IgAN+Antroq小鼠之腎組織病理仍再度受到 顯著抑制。 腎織維化相關基因及蛋白質表現 83 201247612 在此,係檢測小鼠中TGF-βΙ與Co丨-IV之mRNA及蛋白 質表現量。雖然AcP-IgAN控制組小鼠,相較於正常控制組 小鼠,其TGF-βΙ與Col-IV之mRNA表現在第28天時均大幅增 加,但在投予化合物1之AcP-IgAN+Antroq小鼠,此現象係 大幅受到抑制,如圖10A-10D所示。在第3天時,僅有兩組 小鼠有觀察到基礎的亮種纖維化相關基因之mRNA基礎表 現量。反之,在第28天時,IHC結果顯示,AcP-IgAN控制 組小鼠之TGF-βΙ與Col-IV之蛋白量係大幅增加,但此現象 在AcP-IgAN+Antroq小鼠中係明顯受到抑制。 鉍胞免疫及腎發炎 在此,係以IgAN病理實驗觀察細胞相關免疫現象。以 脾臟細胞(splenocyte)之流式細胞儀試驗,以分別檢測 CD3、CD4及CD8活性。如圖11A-11C所示,在第3天時,相 較於正常控制組小鼠,AcP-IgAN控制組小鼠即可觀察到 CD3+/CD69+、CD4+/CD69+或 CD8+/CD69+百分比有明顯增 加,但在第28天時仍未觀察到T細胞所有亞型有類似情形發 生。反之,於第3天時,相較於AcP-IgAN控制組小鼠, AcP-IgAN+Antroq小鼠中,以化合物1處理可誘導 CD3+/CD69+T細胞百分比明顯下降。然而,在第3天時, AcP-IgAN控制組小鼠與AcP-IgAN+Antroq小鼠間之 CD4+/CD69+或CD8+/CD69+T細胞百分比則無顯著差異。於 正常控制組小暑中,三種T細胞之每一種亞型百分比,在第 28天仍無險處差異。胸嘴咬吸收分析(thymidine uptake analysis)結果顯示,於第3天時,AcP-IgAN控制組小鼠之脾 84 201247612 臟細胞中CD3+T細胞較AcP-IgAN+Antroq小鼠大幅增生(如 圖11A-11C所示),且在第28天時,每一組小鼠之增生基線 值均與正常控制組小鼠相近。此外,亦透過IHC試驗評估小 鼠腎臟中浸潤之單核白血球細胞表現。如圖12A-12F所示, 相較於正常控制組小鼠,AcP-IgAN小鼠雖然在第3天時僅觀 察到腎臟申有少量發炎細胞,但在第28天時,可在腎臟間 質組織中觀察到局部但明顯的CD4+ Th細胞、CD8+ Tc細 胞、CDllc+噬中性白血球、F4/80+單核細胞/巨噬細胞、及 CDllb+單核細胞/巨嗟細胞《相反的,相較於Acp_igAN小 鼠,於第28天時,AcP-IgAN+Antroq小鼠之腎臟之發炎細胞 浸潤有顯著減少,且在第3天時,AcP-IgAN+Antroq小鼠之 腎臟幾乎無法檢測到pan-T細胞、嗟中性白血球細胞、及單 核白血球/巨噬細胞之訊號。 氧化壓力、Nrf2及相關路徑 ROS係為判定各種形式腎病(包括igAN)之加速或惡化 的主要有害化學媒介之一。在血液可系統性的檢測到ROS 表現量’而在腎臟組織可局部觀察到ROS表現量。相較於 正常控制組小鼠,AcP-IgAN小鼠在第3天及第28天時,血清 中有顯著R0S量增加,而在尿液及腎臟組織中,則在第28 天時觀察到ROS量增加’如圖13A-13F所示。相反的,相較 於AcP-IgAN控制組小鼠,投予本發明示例化合物1之 AcP-IgAN+Antroq小鼠,於第3天時,其血清中之R〇S量有 大量減少,且在第28天時’其血清、尿液及腎臟組織之r〇s 量也大量減少。此外,在第3天直到第28天,AcP-IgAN控制 85 201247612 組小鼠之尿液一氧化氮(NO)量均高於正常控制組小鼠。然 而,相較於AcP-IgAN控制組小鼠,AcP-IgAN+Antroq小鼠 於第3天及第28天時,均可觀察到顯著NO量減少。雖然 AcP-IgAN控制組小鼠與AcP-IgAN+Antroq小鼠在第3天時 血清中NO量無顯著差異,但在第28天時,AcP-IgAN+Antroq 小鼠之血清中NO量較AcP-IgAN小鼠大幅減少。 接著,將投予本發明示例化合物1,以觀察造成上述結 果的可能機轉因素》如圖14A-14F所示,從第3天至小鼠犧 牲之第28天為止,相較於AcP-IgAN控制組小鼠之Nrf2之 mRNA及蛋白質基礎表現量,AcP-IgAN+Antroq小鼠之Nrf2 之mRNA及蛋白質表現量均大幅增加。此外,Nrf2下游蛋白 之榖胱甘肽過氧化酶(GPx),在第28天時,AcP-IgAN+Antroq 小鼠之腎臟組織中的表現量係顯著高於AcP-IgAN控制組小 鼠,雖然此兩組小鼠在第3天時並無明顯差異。 血清促發炎因子量 首先,如圓15B所示,AcP-IgAN控制組小鼠在第3天時 血清MCP-1量有顯著增加,此現象持續到第28天小鼠犧牲 均有觀察到。相反的,在AcP-IgAN+Antroq小鼠中’此現象 係大幅受到抑制,且與基礎量相近。此外,雖然在第3天時 AcP-IgAN控制組小鼠、AcP-IgAN+Antroq小鼠及正常控制 組小鼠之血清IL-6量無顯著差異,但在第28天時’ AcP-IgAN+Antroq小鼠之血清IL-6量有顯著下降(如圖15A 所示),而AcP-IgAN控制組小鼠之血清IL-6量則較正常控制 組小鼠有顯著增加。於第28天時,AcP-IgAN控制組小鼠之 86 201247612 血清IL-Ιβ量較正常控制組小鼠明顯增加,但此現象在 AcP-IgAN+Antroq小鼠中係明顯受到抑制(如圖15C所示)。 於第3天時,無論是AcP-IgAN控制組小鼠及 八〇?-4八1<1+六111^〇9小鼠之11^-18量,均未觀察到較正常控制 組小鼠有增加的現象,如圖15D所示。雖然相較於正常控制 組小鼠,AcP-IgAN控制組小鼠與AcP-IgAN+Antroq小鼠在 第3天時之血清IL-18量增加幾乎相同,但於第28天時, AcP-IgAN控制組小鼠之血清IL-18量貝'J明顯提升,而在 AcP-IgAN+Antroq小鼠中,血清IL-18量則受到抑制。 NLRP3發炎逋活化(腎臟中) 含NACHT、LRR及PYD區域蛋白3 (NALP3)發炎艎係與 發炎反應有高度相關,且與適應性免疫(adaptive immunity) 有關"雖然NLRP3於主體反應及病理相關分子間關係已受 到證實,但其與免疫複合相關腎絲球疾病間的關係仍尚不 清楚"無論NALP3發炎活化是否會在AcP-IgAN小鼠中產 生,AcP-:[gAN小鼠模型的建立,仍與腎臟局部性發炎反應 增加有關。在第3天及第28天,雖然AcP-IgAN小鼠較正常控 制組小鼠之腎贜中有明顯NLRP3蛋白質增加,但此現象在 AcP-IgAN+Antroq小鼠中係明顯受到抑制。在AcP-IgAN控 制組小鼠及AcP-IgAN+Antroq小鼠中,NLRP3之mRNA表現 量均較正常控制組小鼠高,但投予化合物1並未對 AcP-IgAN+Antroq小鼠之NLRP3之mRNA表現有所影響。然 而,在第28天時,AcP-IgAN+Antroq小鼠之腎臟NLRP3之 mRNA表現較AcP-IgAN控制組小鼠顯著減少,其中 87 201247612The IgAN model was administered with AcP-IgAN mice (AcP-IgAN control group mice) as a disease control group, which increased the amount of urinary protein on the 7th day of the induced disease and continued until the 28th day after the end of the experiment. Rise, as shown by circle 9. However, AcP-IgAN mice (AcP-IgAN + Antroq mice) treated with Compound 1 were greatly inhibited, although some micro-urinary protein production was observed compared to the normal control group. . In addition, the AcP-IgAN control group had a significant increase in BUN serotype (Fig. 9B) and serum creatinine (Fig. 9C) on day 28; although AcP-IgAN+Antroq mice and AcP-IgAN control group, normal Group mice did not have significant differences in BUN and Cr serum levels on day 3, but AcP-IgAN+Antroq mice showed better renal function. 82 201247612 The growth of the mice in the eAcP-IgAN+Antroq mice and the AcP-IgAN control group mice was measured weekly, which was no different from the normal control group. In addition, all mice in each group exhibited normal activity without a change in hair loss or appetite. Renal pathology tests are shown in Figures 9A-9E. On day 28, mice in the AcP-IgAN control group had neutrophil infiltration, local but typical crescent, and/or spheroidal segmental sclerosis. Correlation of increased infiltration, and significant renal mononuclear leukocyte infiltration and scattered tubular atrophy (related to protein shedding), this result shows that compared with the previous study of renal spheroid tissue pathology report (Lai, KN Nephron. 92:263-270 (2002) ; Lai, et al., Nephron. 69:1-8 (1995); Chen, et al., J. Clin. Lab. Analysis. 6:35-39 (1992) Kashem, et al". /«ί· 45: 868-875 (1994)), and histopathological study of the renal interstitial (Falk, et al., along the curry/«ί. 47:177-185 (1995) Van Es, et al., Kidney Int. 73:1426-1433 (2008); Torres, et al., Kidney Int. 73:327-333 (2008); Walsh, et al., Clin J Am Soc. Nephrol 5: 425-430 (2010); Fujinaka, et al., J. #ep;7rc)/· 20:357-363 (2007)), with an increased and worsened state of kidney disease, except for renal glomerular immunoprecipitation All other kidney damage in AcP-IgAN+Antroq mice, other than kidney damage All of them were inhibited. On the third day, the control group AcP-IgAN mice began to have localized nephropathy, but the renal histopathology of AcP-IgAN+Antroq mice treated with Compound 1 was again significantly inhibited. Renal DNA-related genes and protein expression 83 201247612 Here, the mRNA and protein expressions of TGF-βΙ and Co丨-IV in mice were detected. Although the AcP-IgAN control group mice were smaller than the normal control group. In mice, the mRNA expression of TGF-βΙ and Col-IV increased significantly on the 28th day, but in the AcP-IgAN+Antroq mice administered Compound 1, this phenomenon was greatly inhibited, as shown in Figures 10A-10D. On the third day, only two groups of mice observed the underlying mRNA expression of the basal bright fibrosis-related genes. Conversely, on day 28, the IHC results showed that the AcP-IgAN control group mice The amount of protein of TGF-βΙ and Col-IV was greatly increased, but this phenomenon was significantly inhibited in AcP-IgAN+Antroq mice. Cellular Immunity and Renal Inflammation Here, cell-associated immune phenomena are observed by IgAN pathology experiments. Flow cytometry experiments with splenocytes were performed to detect CD3, CD4 and CD8 activities, respectively. As shown in Figures 11A-11C, on day 3, a significant increase in the percentage of CD3+/CD69+, CD4+/CD69+ or CD8+/CD69+ was observed in the AcP-IgAN control group compared to the normal control group. However, no similar situation occurred in all subtypes of T cells on day 28. Conversely, on day 3, treatment with Compound 1 induced a significant decrease in the percentage of CD3+/CD69+ T cells in AcP-IgAN+Antroq mice compared to AcP-IgAN control group mice. However, on day 3, there was no significant difference in the percentage of CD4+/CD69+ or CD8+/CD69+ T cells between AcP-IgAN control group mice and AcP-IgAN+Antroq mice. In the normal control group, the percentage of each of the three T cells was not different on the 28th day. The results of thymidine uptake analysis showed that on day 3, CD3+ T cells in the spleen 84 201247612 spleen cells of the AcP-IgAN control group were significantly more proliferating than AcP-IgAN+Antroq mice (Fig. 11A-11C), and on day 28, the baseline values of proliferation in each group of mice were similar to those in the normal control group. In addition, mononuclear white blood cell performance infiltrated in the kidneys of the mice was also assessed by the IHC test. As shown in Figures 12A-12F, AcP-IgAN mice observed only a small amount of inflammatory cells in the kidney on day 3, but on the 28th day, in the renal interstitial. Local but significant CD4+ Th cells, CD8+ Tc cells, CDllc+ phagocytic leukocytes, F4/80+ monocytes/macrophages, and CD11b+monocytes/macrophage cells were observed in the tissues. In Acp_igAN mice, the inflammatory cell infiltration of the kidneys of AcP-IgAN+Antroq mice was significantly reduced on day 28, and on day 3, the kidneys of AcP-IgAN+Antroq mice were almost unable to detect pan-T. Signals for cells, sputum neutrophils, and mononuclear leukocytes/macrophages. Oxidative stress, Nrf2, and related pathways ROS is one of the major harmful chemical mediators for determining the acceleration or worsening of various forms of kidney disease, including igAN. The amount of ROS expression can be systematically detected in the blood and the amount of ROS expression can be observed locally in the kidney tissue. Compared with the normal control group, AcP-IgAN mice showed a significant increase in serum ROS on day 3 and day 28, while in urine and kidney tissues, ROS was observed on day 28 The amount increase is as shown in Figures 13A-13F. In contrast, AcP-IgAN+Antroq mice administered the exemplified compound 1 of the present invention had a large decrease in the amount of R〇S in the serum on day 3 as compared with the AcP-IgAN control group mice. On day 28, the amount of r〇s in serum, urine and kidney tissue was also greatly reduced. In addition, on day 3 until day 28, the amount of urinary nitric oxide (NO) in the AcP-IgAN control 85 201247612 group was higher than that in the normal control group. However, a significant decrease in NO was observed on days 3 and 28 of AcP-IgAN + Antroq mice compared to AcP-IgAN control mice. Although there was no significant difference in the amount of NO in serum between AcP-IgAN control group and AcP-IgAN+Antroq mice on day 3, at day 28, the amount of NO in the serum of AcP-IgAN+Antroq mice was higher than that of AcP. -IgAN mice were substantially reduced. Next, the exemplified compound 1 of the present invention will be administered to observe the possible mechanism for causing the above results, as shown in Figs. 14A to 14F, from the third day to the 28th day of the sacrifice of the mouse, compared to AcP-IgAN. The mRNA and protein base expression of Nrf2 in the control group mice, and the mRNA and protein expression of Nrf2 in AcP-IgAN+Antroq mice were greatly increased. In addition, the glutathione peroxidase (GPx) of the downstream protein of Nrf2, on day 28, the expression level in the kidney tissue of AcP-IgAN+Antroq mice was significantly higher than that in the AcP-IgAN control group, although There was no significant difference between the two groups of mice on day 3. Amount of serum proinflammatory cytokines First, as shown by circle 15B, the amount of serum MCP-1 was significantly increased in the AcP-IgAN control group on day 3, and this phenomenon continued until the 28th day of mouse sacrifice. In contrast, in AcP-IgAN + Antroq mice, this phenomenon was greatly suppressed and was similar to the basal amount. In addition, although there was no significant difference in serum IL-6 levels between AcP-IgAN control group mice, AcP-IgAN+Antroq mice, and normal control group mice on day 3, on day 28, 'AcP-IgAN+ The amount of serum IL-6 in Antroq mice was significantly decreased (as shown in Figure 15A), while the serum IL-6 levels in the AcP-IgAN control group were significantly higher than those in the normal control group. On day 28, the amount of serum IL-Ιβ in the 86 201247612 mice in the AcP-IgAN control group was significantly higher than that in the normal control group, but this phenomenon was significantly inhibited in AcP-IgAN+Antroq mice (Fig. 15C). Shown). On day 3, no more normal control group mice were observed in the AcP-IgAN control group mice and the 11^-18 doses of the gossip--4 八1<1+6111^〇9 mice. There is an increase phenomenon as shown in Fig. 15D. Although the amount of serum IL-18 in the AcP-IgAN control group and AcP-IgAN+Antroq mice increased almost the same on day 3 compared to the normal control group, on day 28, AcP-IgAN Serum IL-18 levels in control mice were significantly increased, whereas in AcP-IgAN + Antroq mice, serum IL-18 levels were inhibited. NLRP3 inflammatory 逋 activation (in the kidney) NACHT, LRR and PYD region protein 3 (NALP3) inflammatory sputum is highly correlated with inflammatory response and is associated with adaptive immunity" although NLRP3 is associated with subjective and pathological changes Intermolecular relationships have been confirmed, but its relationship with immune complex-associated renal glomerular disease remains unclear. "Whether NALP3 inflammatory activation will occur in AcP-IgAN mice, AcP-: [gAN mouse model Established, still associated with increased local inflammation of the kidney. On day 3 and day 28, although AcP-IgAN mice had a significant increase in NLRP3 protein in the renal pelvis of the normal control group, this phenomenon was significantly inhibited in AcP-IgAN + Antroq mice. In the AcP-IgAN control group and AcP-IgAN+Antroq mice, the mRNA expression of NLRP3 was higher than that in the normal control group, but compound 1 was not administered to NLRP3 of AcP-IgAN+Antroq mice. mRNA performance has an effect. However, on day 28, the mRNA expression of NLRP3 in the kidney of AcP-IgAN+Antroq mice was significantly lower than that in the AcP-IgAN control group, 87 87476612

AcP-IgAN控制组小鼠之此基因之腎臟niRNA表現量仍較正 常控制組小鼠高許多。重要的是,在第3天至第28天時,由 定量即時PCR分析結果顯示,AcP-IgAN+Antroq小鼠之凋亡 因子-1及IL-18之腎臟mRNA表現量均大幅增加,但此險象 在AcP-IgAN+Antroq小鼠均受到抑制,如圖16A-16F所示》 此外,在第3天及第28天,相較於AcP-IgAN控制組小鼠,投 予Antroq之AcP-IgAN+Antroq小鼠之腎臟凋亡因子·1蛋白 質表現量係受到抑制,雖然第28天時AcP-IgAN+Antroq小鼠 之腎IL-18蛋白產生係明顯受到抑制。於第28天時, AcP-IgAN控制組小鼠之IL-Ιβ之mRNA表現量有大幅提 升,但在AcP-IgAN+Antroq小鼠中卻受到顯著抑制,雖然在 第3天時並未觀察到腎臟IL-Ιβ之mRNA表現量有所增加。 腎NF-κΒ活化及其相關細胞因子 基於AcP-IgAN小鼠腎臟中明顯的單核白血球細胞浸 潤現象,腎贜中局部發炎反應似乎為IgAN加速及發展的主 要路徑。在此,係研究NF-κΒ在腎臟中所扮演的角色。首先, 如圖17A-17F所示,相較於AcP-IgAN控制組小鼠(其在第28 天時腎臟中核NF-κΒ蛋白量較正常控制組小鼠顯著增加), AcP-IgAN+Antrqo小鼠腎臟中之核NF-kB蛋白表現量係顯 著減少,雖然在第3天時,無論是AcP-IgAN控制組小鼠或 AcP-IgAN+Antrqo小鼠之此蛋白腎臟中表現量均與正常控 制組小鼠差異不大。此外’由第28天之腎臟組織IHC結果顯 示,相較於AcP-IgAN控制組小鼠有顯著的核轉移增加’化 合物1更可具有顯著減少AcP-IgAN+Antrqo小鼠之NF-κΒ核 88 201247612 轉移之功效。此外,更分別進行腎臟中MCP-1 (圖17C)及IL-6 (圖17D)之mRNA與蛋白質表現的定量分析。雖然AcP-AgNA 控制組小鼠、AcP-IgAN+Antrqo小鼠與正常控制組小鼠在第 3天時結果無明顯差異,但於第28天時,相較於AcP-IgAN 控制組小鼠,投予化合物1可明顯減少AcP-IgAN+Antrqo小 鼠腎臟中MCP-1及IL-6的mRNA及蛋白質表現量,如圖17E 及圖17F所示。 腎臟中細胞凋亡 腎臟中之細胞凋亡試驗係用以了解IgAN之病理。如圖 18八-188所示,1'13\五1^實驗結果顯示,八〇?-1§八1^控制組在 第28天時,其細胞凋亡情形較正常控制組小鼠顯著增加, 但此現象在投予Antroq之AcP-IgAN+Antroq小鼠中卻明顯 受到抑制;即便在第3天時所有小鼠之細胞凋亡現象均不明 顯。 實施例15 :腸道外投藥劑型 製備適合用於腸道外注射投藥之醫藥組成物,係將100 mg之本發明所述之化合物或其鹽類溶於dmsO中,而後與 10 mL之0.9%無菌生理時鹽水混合。而後,將此混合物係包 裝在一適用於注射投藥之藥物單元中。 實施例16: 口服劑型 製備用於口服投藥之醫藥組成物,係將1〇〇 mg之示範 化合物1與1〇〇 mg之玉米油混合。而後,將此混合物係包裝 在一口服單元中,如膠囊中,以適用於口服投藥。 89 201247612 於部分實施例中,100 mg之本發明所述之化合物係與 750 mg之澱粉混合,並將此混合物係包裝在一口服單元 中,如硬質的明膠膠囊中,以適用於口服投藥。 實施例17:舌下投藥(硬質錠劑)劑型 製備用以口頰投藥之醫藥組成物,如硬質錠劑劑型, 係將100 mg之本發明所述之化合物與420 mg糖粉末混合, 再與1.6 mL之淡玉米糖漿、2.4 mL蒸餾水、及0.42 mL薄荷 萃取物混合。而後,小心研磨混合物,並將研磨後之混合 物倒入一模型中,以形成適用於口頰投藥之錠劑= 實施例18 :吸入组成物 製備用以吸入式投藥之醫藥組成物,係將20 mg之本發 明所述之化合物與50 mg之無水擦檬酸及100 mL之0.9%氣 化鈉溶液混合。而後,將此混合物係包裝在一吸入式投藥 單元中,如喷霧氣,以適用於吸入式投藥。 實施例19 :直腸凝膠劑型 製備用以直腸投藥之醫藥組成物,係將100 mg之本發 明所述之化合物與2.5 g之甲基纖維素(1500 mPa)、100 mg 之甲基對苯甲酸(methylparapen)、5 g之甘油、及100 mL之 純水混合。而後,將將所得到之凝膠混合物包裝在一直腸 投藥單元中,如注射器,以適用於直腸投藥。 實施例20:外用凝膠組成物The renal niRNA expression of this gene in the AcP-IgAN control group mice was still much higher than that in the normal control group. Importantly, from day 3 to day 28, quantitative real-time PCR analysis showed that the mRNA expression levels of apoptotic factor-1 and IL-18 in AcP-IgAN+Antroq mice increased significantly, but this The risk was inhibited in AcP-IgAN+Antroq mice, as shown in Figures 16A-16F. In addition, on days 3 and 28, Antroq's AcP-IgAN was administered compared to the AcP-IgAN control group. The renal apoptotic factor-1 protein expression was inhibited in +Antroq mice, although the renal IL-18 protein production line of AcP-IgAN+Antroq mice was significantly inhibited on day 28. On day 28, the mRNA expression of IL-Ιβ in the AcP-IgAN control group was significantly increased, but it was significantly inhibited in AcP-IgAN+Antroq mice, although it was not observed on the third day. The mRNA expression of IL-Ιβ in the kidney increased. Renal NF-κΒ activation and related cytokines Based on the apparent mononuclear leukocyte infiltration in the kidneys of AcP-IgAN mice, local inflammatory response in the renal pelvis appears to be the main pathway for IgAN acceleration and progression. Here, we study the role of NF-κΒ in the kidney. First, as shown in Figures 17A-17F, compared to the AcP-IgAN control group mice (the amount of nuclear NF-κΒ protein in the kidney was significantly increased on day 28 compared with the normal control group), AcP-IgAN+Antrqo was small. The nuclear NF-kB protein expression in the rat kidney was significantly reduced, although on day 3, both the AcP-IgAN control group or the AcP-IgAN+Antrqo mouse had a normal expression of the protein in the kidney. The mice in the group did not differ much. In addition, the IHC results from kidney tissue on day 28 showed a significant increase in nuclear transfer compared to the AcP-IgAN control group. Compound 1 was more able to significantly reduce NF-κ nucleus 88 in AcP-IgAN+Antrqo mice. 201247612 The effect of the transfer. In addition, quantitative analysis of mRNA and protein expression of MCP-1 (Fig. 17C) and IL-6 (Fig. 17D) in the kidneys was performed. Although the AcP-AgNA control group mice, AcP-IgAN+Antrqo mice and the normal control group mice did not differ significantly on the third day, on the 28th day, compared with the AcP-IgAN control group mice, Administration of Compound 1 significantly reduced the mRNA and protein expression of MCP-1 and IL-6 in the kidney of AcP-IgAN+Antrqo mice, as shown in Figures 17E and 17F. Apoptosis in the kidney The apoptosis test in the kidney is used to understand the pathology of IgAN. As shown in Fig. 18-188, the results of 1'13\5 1^ showed that the apoptosis of the control group at the 28th day was significantly higher than that of the normal control group. However, this phenomenon was significantly inhibited in AcP-IgAN+Antroq mice administered to Antroq; even on day 3, apoptosis was not observed in all mice. Example 15: Parenteral dosage form A pharmaceutical composition suitable for parenteral injection administration is prepared by dissolving 100 mg of the compound of the present invention or a salt thereof in dmsO, followed by 0.9% sterility with 10 mL. Mix saline when physiological. The mixture is then packaged in a pharmaceutical unit suitable for injectable administration. Example 16: Oral dosage form A pharmaceutical composition for oral administration was prepared by mixing 1 mg of the exemplary compound 1 with 1 mg of corn oil. The mixture is then packaged in an oral unit, such as a capsule, for oral administration. 89 201247612 In some embodiments, 100 mg of the compound of the invention is mixed with 750 mg of starch and the mixture is packaged in an oral unit, such as a hard gelatin capsule, for oral administration. Example 17: Sublingual Administration (Hard Tablets) Formulations A pharmaceutical composition for buccal administration, such as a hard tablet dosage form, is prepared by mixing 100 mg of a compound of the invention with 420 mg of sugar powder, and Mix 1.6 mL of light corn syrup, 2.4 mL distilled water, and 0.42 mL of peppermint extract. Thereafter, the mixture is carefully ground and the ground mixture is poured into a mold to form a lozenge suitable for buccal administration = Example 18: Inhalation Composition Preparation of a pharmaceutical composition for inhalation administration, system 20 The compound of the invention of mg is mixed with 50 mg of anhydrous citric acid and 100 mL of a 0.9% sodium hydride solution. The mixture is then packaged in an inhalation dosing unit, such as a spray gas, for inhaled administration. Example 19: Rectal Gel Formulation A pharmaceutical composition for rectal administration is prepared by applying 100 mg of the compound of the present invention to 2.5 g of methylcellulose (1500 mPa), 100 mg of methylparabenzate. (methylparapen), 5 g of glycerin, and 100 mL of pure water. The gel mixture obtained will then be packaged in a continuous enteral administration unit, such as a syringe, for rectal administration. Example 20: Topical gel composition

製備外用凝膠醫藥組成物,係將100 mg之本發明所述 之化合物與1.75 g之羥丙基纖維素、10 mL之丙二醇、10 mL 之肉豆 Μ 酸異丙 S 旨(isopropyl myristate)、及 100 mL 之 USP 90 201247612 級乙醇。而後,將所製得之凝膠混合物包裝在一容器中, 如軟管’以適用於以外用方式投藥。 實施例21 :眼藥水组成物 製備眼藥水組成誤,係將100 mg之本發明所述之化合 物與〇.9g之Nac丨(溶於l〇〇mL純水中)混合,並使用〇2微米 過濾膜進行過濾。而後,將所得之非離子性溶液包裝在一 眼部投藥單元中,如眼藥水容器,以適用於透過眼部進行 投藥&quot; 本發明雖揭示且描述較佳實施例,但本技術領域者均 知道這些實施例僅用以說明用。本技術領域者可在不偏離 本發明之範圍下,進行各種改變、修飾、及取代。此外, 對本發明實施例所做的各種改變,均應解釋為用以實施本 發明。同時,本發明所主張之權利範圍自應以申請專利範 圍所述為準’且本發明申請專利範圍所主張之方法及結構 及其均等物,均涵蓋於本發明之範圍中。 【圖式簡單說明】 本發明之特徵係如申請專利範圍所述。本發明所提供 之實施例係用以詳述本發明之發明特徵,且藉由參考本發 明實施例及圖式’可更佳了解本發明之特徵及優點。 圖1A-1C係為不例環己烯酮化合物丨抑制尿蛋白及提升腎功 能之、.·〇果.(圖1A)尿蛋白隨時間變化之研究;(圖1B)血 /月中血尿素氣(BUN)含量;(圖ic)血清肌酸酐量,其中, 201247612 此些數據係以每組各6隻小鼠所得到之實驗平均值士SEM表 示,其中 *;?&lt; 0.05、**/?&lt; 0.01、***/?&lt;0.005、#無法债測。 圖2A-2B係為示例化合物1之腎臟組織病理發展抑制結果: (圖2A)於投藥第7、14、21天之H&amp;E染色腎臟組織病理評 估;(圖2B)於投藥第7、14、21天之肌間線蛋白之組織免 疫染色測量腎絲球之足戲胞損傷;其中黑色箭頭前端、白 色箭頭前端、及箭頭分別表示上皮增生損傷(EPHL)、硬化、 及足細胞;最初放大倍率為400X,半定量分析顯示於圓右 側,此些數據係以每組各6隻小鼠所得到之實驗平均值 士SEM表示,其中 *尸&lt; 0.05、**/?&lt; 0.01、***/?&lt;0.005、#無法 偵測。 圖3A-3F係為示例化合物1保護FSGS小鼠生成ROS/NO之結 果:(圖3A)血清中超氧陰離子量;(圖3B)血清中NO量; (圖3C)尿液中超氧陰離子量;(圖3D)尿液中NO量;(圖3E) 腎臟蛋白中超氧陰離子量;(圖3F)二氫乙錠(DHE)標定之 腎臟原位ROS生成;最初放大倍率為400X,半定量分析顯 示於圖右側,此些數據係以每組各6隻小鼠所得到之實驗平 均值土SEM表示,其中 *p&lt; 0.05、**户&lt; 0.01、***/?&lt;0.005。 圖4A-4E係為示例化合物1之提升核Nrf2表現及抑制胞内 P47ph°x蛋白表現之結果:(圖4A)及(圖4B)係分別為胞内 p47ph°x及腎臟組織中Nrf2之西方墨點試驗結果,其中β-肌動 蛋白與組蛋白Η3係分別作為胞内及核蛋白之内控制組;(圖 4C) ρ47ρΗ°χ/β-肌動蛋白比例量化數值;(圖4D) Nrf2/組蛋白 H3比例量化數值;(圖4E)腎臟中GPx活性;此些數據係以 92 201247612 每組各6隻小鼠所得到之實驗平均值士SEM表示,其中 0.05 ' **p&lt; o.oi . ***p&lt;〇.〇〇5 〇 圖5 A - 5 Β係為示例化合物丨之τ細胞及巨噬細胞浸潤結果 圖:(圖5Α)以組織免疫染色法測量CD3+ τ細胞結果;(圖 5B)以組織免疫染色法測量F4/8〇單核白血球/巨噬細胞結 果,最初放大倍率為4〇〇χ,紅色箭頭顯示CD3+ τ細胞,半 疋量分析顯示於圖右側,且此些數據係以每組各6隻小鼠所 得到之實驗平均值:fcSEM表示,其中〇.〇5、**p&lt; 〇 〇1、 ***;?&lt;0.005 » 圓6A-6C係為示例化合物j之抑制腎臟中IL 6表現 活性之結果:(圖6A)以組織免疫染色法測量江_6蛋白;(圓 6B)以組織免疫染色法測量測量ΝΙγ·κΒ p65,其中最初放大 倍率為400X,半定量分析顯示於圖右側;(圖6C) 活性;此些數據係以每組各6隻小鼠所得到之實驗平均值 士SEM表示’其中 〇.05、*&gt;&lt; 〇 〇1、⑻5。 圖7A-7C係為示例化合之抑制腎臟中膠原蛋ΑΙ πι&amp;ιν 聚集之結果:(圖7Α)以免疫組織染色偵測膠原蛋白1;(圖 7Β)以免疫組織染色偵測膠原蛋白111;(圖7C)以免疫組織 染色偵測膠原蛋白IV ;最初放大倍率為4〇〇χ,半定量分析 顯示於圖右側,此些數據係以每組各6隻小鼠所得到之實驗 平均值±SEM表示,其中 **ρ&lt; 〇.〇1、。 圖8A-8C係為示例化合物1之抑制血清及腎臟组織中 TGF-βΙ表現之結果:(圖8Α)以免疫組織染色偵測血清中 TGF-βΙ量;(圖8Β)以免疫組織染色偵測腎蛋白中^ 93 201247612 量;(圖8C)以免疫組織染色偵測TGF-β 1 ;最初放大倍率為 400X,半定量分析顯示於圖右側,此些數據係以每組各6 隻小鼠所得到之實驗平均值土 SEM表示,其中 0.01、***p&lt;〇.005。 圖9A-9E係為示例環己烯酮化合物1之減少AcP-IgAN小鼠 中尿蛋白及提升腎功能且作為腎臟組織病理切片之結果: (圖9A)尿蛋白隨時間變化之研究;(圖9B)血清中血尿素 氮(BUN)含量;(圖9C)血清肌酸酐量;(圓9D)於第3天及 第28天處理後’以h&amp;e染色評估腎臟之組織病理;(圖9e) 於第3天及第28天處理後,以PAS染色評估腎臟之組織病 理;最初放大倍率為400X,受影響的腎絲球百分比係列於 圖下方,此些數據係以每組各6隻小鼠所得到之實驗平均值 ±SEM表示,其中 *p&lt; 0.05、**p&lt; 0.01、〇〇5、#無法 债測。 圓10A-10D係為以示例環己稀辆化合物1餵食Acp_igAN小 鼠時TGF- βΐ及Col-IV之mRNA與蛋白質表現量結果;(圓 10A)以即時PCR偵測TGF-βΙ於腎臟中之mRNA表現量結 果;(圖10B)以即時PCR偵測膠原蛋白I於腎臟中之mRNA 表現量結果;(圖10C)以免疫組織染色偵測TGF-βΙ於腎臟 中之蛋白質表現量結果;(圖10D)以免疫組織染色偵測膠 原蛋白I於腎臟中之蛋白質表現量結果;最初放大倍率為 400X’數值係列於圖下方,此些數據係以每組各6隻小鼠所 得到之實驗平均值士SEM表示,其中**;?&lt; 〇.(H、***p&lt;〇.〇〇5。 94 201247612 圖1 ΙΑ-11C係為以流式細胞儀偵測脾臟細胞中IgAN病理之 細胞相關免疫反應之結果:(圖11A)第3天及第28天處理後 之CD3 +脾臟細胞中CD3+CD69+細胞百分比;(圓11B)第3 天及第28天處理後之CD19+脾臟細胞中CD19+CD69+細胞百 分比;(圖11C) T細胞增生結果;此些數據係以每組各6隻小 鼠所得到之實驗平均值士SEM表示,其中*/?&lt; 〇.〇5、**p&lt; 0.01、**&gt;&lt;〇.〇〇5、#無法偵測。 圖12A-12F係為AcP-IgAN小鼠腎臟中單核白金球細胞浸调 之型態表現評估結果:(圊12A)以免疫螢光染色測量CD3 + T細胞結果;(圖12B)以免疫螢光染色測量CD4+ T細胞結 果;(圖12C)以免疫螢光染色測量CD8+ T細胞結果;(圖12d) 以免疫組織染色測量CD1 lb巨噬細胞/噬中性白血球細胞結 果;(圖12E)以免疫組織染色測量CDl lc樹突細胞結果;(圖 12F)以免疫組織染色測量F4/80單核細胞/巨噬細胞結果; 最初放大倍率為400X,數值係列於圖下方,此些數據係以 每組各6隻小鼠所得到之實驗平均值±SEM表示,其中*p&lt; 0.05、**/&gt;&lt; 0.01、***;?&lt;〇.〇〇5、#無法偵測。 圖13A-13F係為示例環己稀嗣化合物1抑制AcP-IgAN小鼠 中ROS/NO產生之結果:(圖13A)血清中超氧陰離子量;(圖 13B)血清中NO量;(圖13C)尿液中超氡陰離子量;(圖13D) 尿液中NO量;(圖13E)腎臟中超氧陰離子量;(圖13F)以二 氫乙錠(DHE)染色得到之腎臟原位R〇s產生偵側結果;最初 放大倍率為400X ’數值係列於圖右側,此些數據係以每組Preparation of a topical gel pharmaceutical composition comprising 100 mg of a compound of the invention with 1.75 g of hydroxypropylcellulose, 10 mL of propylene glycol, 10 mL of isopropyl myristate, And 100 mL of USP 90 201247612 grade ethanol. The gel mixture thus prepared is then packaged in a container, such as a hose, for administration in a suitable manner. Example 21: Eye Drop Composition The eyedrop composition error was prepared by mixing 100 mg of the compound of the present invention with 〇.9 g of Nac丨 (dissolved in 100 mL of pure water) and using 〇2 μm. The filter membrane was filtered. The resulting non-ionic solution is then packaged in an ocular administration unit, such as an eye drop container, for application to the eye. <Desc/Clms Page number>> These examples are understood to be illustrative only. Various changes, modifications, and substitutions can be made by those skilled in the art without departing from the scope of the invention. In addition, various changes to the embodiments of the invention are to be construed as the invention. At the same time, the scope of the claims of the present invention is intended to be within the scope of the present invention. BRIEF DESCRIPTION OF THE DRAWINGS The features of the present invention are as set forth in the claims. The embodiments of the present invention are set forth to provide a better understanding of the features and advantages of the present invention. Figure 1A-1C is a study of the inhibition of urinary protein and the improvement of renal function by a cyclohexenone compound. (Fig. 1A) Study of urine protein over time; (Fig. 1B) Blood/month blood urea Gas (BUN) content; (Figure ic) serum creatinine amount, wherein, 201247612 These data are expressed by the mean SEM of each group of 6 mice, wherein *;?&lt; 0.05, ** /?&lt;0.01, ***/?&lt;0.005,#Unable to measure. 2A-2B are the results of inhibition of renal histopathological development of the exemplified compound 1: (Fig. 2A) H&E staining renal histopathological evaluation on days 7, 14, and 21 of administration; (Fig. 2B) on administration of drugs 7, 4 Tissue immunostaining of 21-day intermuscular line protein was used to measure the cerebral injury of the glomerulus of the kidney; the front of the black arrow, the front end of the white arrow, and the arrow indicate epithelial hyperplasia (EPHL), sclerosis, and podocytes, respectively; The magnification was 400X, and the semi-quantitative analysis was shown on the right side of the circle. These data were expressed as the mean SEM of each group of 6 mice, of which corpse &lt; 0.05, **/?&lt; 0.01, * **/?&lt;0.005,# cannot be detected. 3A-3F are the results of exemplified compound 1 to protect FSGS mice from ROS/NO: (Fig. 3A) the amount of superoxide anion in serum; (Fig. 3B) the amount of NO in serum; (Fig. 3C) the amount of superoxide anion in urine; (Fig. 3D) The amount of NO in the urine; (Fig. 3E) The amount of superoxide anion in the kidney protein; (Fig. 3F) The in situ ROS production of the kidney calibrated with dihydrogen ingot (DHE); the initial magnification is 400X, semi-quantitative analysis shows On the right side of the figure, these data are expressed as the mean SEM of the experimental results obtained for each group of 6 mice, where *p &lt; 0.05, ** household &lt; 0.01, *** /? &lt; 0.005. 4A-4E are the results of the enhanced nuclear Nrf2 expression of the exemplified compound 1 and the inhibition of intracellular P47ph°x protein expression: (Fig. 4A) and (Fig. 4B) are the intracellular p47ph°x and the west of Nrf2 in the kidney tissue, respectively. Ink dot test results, in which β-actin and histone Η3 lines were used as intracellular and nuclear protein control groups, respectively; (Fig. 4C) ρ47ρΗ°χ/β-actin ratio quantified values; (Fig. 4D) Nrf2 /Histone H3 ratio quantified values; (Fig. 4E) GPx activity in the kidney; these data are expressed as SEM of the experimental mean of each of 6 mice in each group of 92 201247612, of which 0.05 ' **p&lt; o. Oi. ***p&lt;〇.〇〇5 〇 Figure 5 A - 5 Β is a sample compound 丨 τ cell and macrophage infiltration results: (Figure 5 Α) measured by tissue immunostaining CD3 + τ cell results; (Fig. 5B) The F4/8 〇 mononuclear leukocyte/macrophage results were measured by tissue immunostaining with an initial magnification of 4 〇〇χ, the red arrow shows CD3+ τ cells, and the half sputum analysis is shown on the right side of the figure. These data are obtained by the average of the experiments obtained for each group of 6 mice: fcSEM, where 〇. 〇5, **p&lt; 〇〇1, ***;?&lt;0.005 » Round 6A-6C is the result of inhibition of IL-6 expression activity in the kidney by the example compound j: (Fig. 6A) measured by tissue immunostaining Jiang_6 protein; (circle 6B) measured by tissue immunostaining measurement of ΝΙγ·κΒ p65, where the initial magnification is 400X, semi-quantitative analysis is shown on the right side of the figure; (Fig. 6C) activity; The experimental mean SEM obtained for 6 mice indicates 'where 〇.05, *&gt;&lt; 〇〇 1, (8) 5. 7A-7C are the results of exemplifying the inhibition of collagen egg ΑΙ πι &amp;ιν aggregation in the kidney: (Fig. 7Α) detecting collagen 1 by immunohistochemical staining; (Fig. 7Β) detecting collagen 111 by immunohistochemical staining; (Fig. 7C) Collagen IV was detected by immunohistochemical staining; the initial magnification was 4 〇〇χ, and the semi-quantitative analysis is shown on the right side of the figure. These data are the mean values of the experiments obtained for each group of 6 mice. SEM indicates that **ρ&lt; 〇.〇1. 8A-8C are the results of inhibiting the expression of TGF-βΙ in serum and kidney tissues of exemplary compound 1: (Fig. 8Α) detecting the amount of TGF-βΙ in serum by immunohistochemical staining; (Fig. 8Β) staining with immune tissue staining In the renal protein, the amount of ^93 201247612 was measured; (Fig. 8C) TGF-β 1 was detected by immunohistochemical staining; the initial magnification was 400X, and the semi-quantitative analysis was shown on the right side of the figure. These data were 6 mice per group. The experimental mean soil SEM obtained is represented by 0.01, ***p &lt; 〇.005. 9A-9E are diagrams showing the reduction of urinary protein and elevation of renal function in AcP-IgAN mice by the cyclohexenone compound 1 and as a result of pathological sectioning of kidney tissue: (Fig. 9A) Study of urine protein changes over time; 9B) blood urea nitrogen (BUN) content in serum; (Fig. 9C) serum creatinine amount; (circle 9D) after treatment on day 3 and day 28 'assessment of renal histopathology by h&e staining; (Fig. 9e After treatment on the 3rd day and the 28th day, the histopathology of the kidney was evaluated by PAS staining; the initial magnification was 400X, and the affected spheroid percentage was below the figure. These data were 6 small each group. The experimental mean ± SEM obtained by the mouse indicates that *p &lt; 0.05, **p &lt; 0.01, 〇〇5, # could not be tested. Round 10A-10D is the result of mRNA and protein expression of TGF-βΐ and Col-IV when feeding Acp_igAN mice with the example cycloheximide compound 1; (circle 10A) to detect TGF-β in the kidney by real-time PCR Results of mRNA expression; (Fig. 10B) Results of mRNA expression of collagen I in the kidney by real-time PCR; (Fig. 10C) Results of protein expression of TGF-β in the kidney by immunohistochemical staining; 10D) The results of protein expression of collagen I in the kidney were detected by immunohistochemical staining; the initial magnification of 400X' is shown below the figure, and the data are the average of the experiments obtained for each group of 6 mice. SEM said that **;?&lt; 〇.(H, ***p&lt;〇.〇〇5. 94 201247612 Figure 1 ΙΑ-11C is a cell for detecting IgAN pathology in spleen cells by flow cytometry Results of related immune responses: (Fig. 11A) Percentage of CD3+CD69+ cells in CD3+ spleen cells after treatment on day 3 and day 28; (circle 11B) CD19+ spleen cells in CD19+ spleen cells after treatment on day 3 and day 28 +CD69+ cell percentage; (Fig. 11C) T cell proliferation results; such data are per The experimental mean SEM of each of the 6 mice was expressed, where */?&lt; 〇.〇5, **p&lt;0.01, **&gt;&lt;〇.〇〇5,# could not be detected. The 12A-12F line was evaluated for the type of mononuclear platinum cell immersion in the kidney of AcP-IgAN mice: (圊12A) to measure CD3 + T cell results by immunofluorescence staining; (Fig. 12B) to immunofluorescence Staining measures CD4+ T cell results; (Fig. 12C) CD8+ T cell results were measured by immunofluorescence staining; (Fig. 12d) CD1 lb macrophage/phagocytic leukocyte results were measured by immunohistochemical staining; (Fig. 12E) immunization Tissue staining was used to measure CD1 lc dendritic cell results; (Fig. 12F) F4/80 monocyte/macrophage results were measured by immunohistochemical staining; initial magnification was 400X, numerical series are below the graph, and these data are in each group. The experimental mean ± SEM obtained for each of the 6 mice, wherein *p &lt; 0.05, **/&gt;&lt; 0.01, ***; ? &lt; 〇.〇〇5, # could not be detected. -13F is an example of cycloheximide compound 1 inhibiting ROS/NO production in AcP-IgAN mice: (Fig. 13A) Superoxide anion in serum; (Fig. 1 3B) the amount of NO in the serum; (Fig. 13C) the amount of superanion in the urine; (Fig. 13D) the amount of NO in the urine; (Fig. 13E) the amount of superoxide anion in the kidney; (Fig. 13F) in dihydroethylene ingot (DHE) The stained kidney in situ R〇s produced a side-detection result; the initial magnification was 400X 'the numerical series is on the right side of the figure, and the data is in each group.

95 201247612 各6隻小鼠所得到之實驗平均值±SEm表示,其中*p&lt; 0.05、 **p&lt; 0.01、***p&lt;〇.〇〇5。 圖14A-14F係為AcP-IgAN小鼠中Nrf2之mRNA及蛋白質表 現量測量結果:(圖14A)以即時PCR測量腎臟Nrf2之mRNA 量之結果;(圏14B)以即時PCR測量腎臟NQ01之mRNA量 之結果;(圖14C)以即時PCR測量腎臟HO-1之mRNA量之結 果;(圖14D)以ELISA測量腎臟中核ΝγΠ表現量之結果;(圖 14Ε)以ELISA測量腎臟中細胞ΗΟ-1表現量之結果;(14F) 腎臟中GPx活性結果;此些數據係以每組各6隻小鼠所得到 之實驗平均值土SEM表示,其中*p&lt; 0.05、**/?&lt; 0.01、 ***尸&lt;〇.〇〇5。 圖15A-15D係為AcP-IgAN小鼠中血清發炎因子量之測量結 果;(圖 15A) IL-6 ;(圖 15B) MCP-1 ;(圖 15C) IL-Ιβ ;(圖 15D) IL-18 ;此些數據係以每組各6隻小鼠所得到之實驗平均值 士SEM表示,其中 *尸&lt; 0.05、**p&lt; 0.01、***;?&lt;〇.〇〇5。 囷16A-16F係為AcP-IgAN小鼠中腎臟NLRP3發炎體活化之 測量結果:(圖16A)以即時PCR測量腎臟NLRP之mRNA表 現量;(圖16B)以即時PCR測量腎臟凋亡因子-1之mRNA表 現量;(圖16C)以即時PCR測量腎臟IL-Ιβ之mRNA表現 量;(圖16D)以即時PCR測量腎臟IL-18之mRNA表現量;(圖 16E)腎臟組織中之NLRP3西方墨點試驗結果;(圖16F)腎 臟組織中之凋亡因子-1 (Casp 1)西方墨點試驗結果;其中, Caspl p20次單元出現代表活化,β-肌動蛋白係作為内控制 組蛋白,0·05、0·01、***ρ&lt;〇·〇〇5。 96 201247612 圖17A-17F係為示例環己烯酮化合物1抑制AcP-IgAN小鼠 腎臟中IL-6與MCP-1表現及增加NF-κΒ活化之結果:(圖17A) 以組織免疫染色測量NF-κΒ p65之結果,其中最初放大倍率 為400X,數值係列於圖下方;(圖17B)使用ELISA-基 TransAM NF-κΒ套組測量腎臟NF-κΒ之結果;(圖17C)以即 時PCR測量腎臟MCP-1之mRNA量之結果;(圖17D)以即時 PCR測量腎臟IL-6之mRNA量之結果;(圖17E)以組織免疫 染色測量腎臟MCP-1之蛋白質量之結果;(圖17F)以組織免 疫染色測量腎臟IL-6之蛋白質量之結果;其中最初放大倍率 為400X,數值係列於圖下方,此些數據係以每組各6隻小鼠 所得到之實驗平均值土SEM表示,其中*/7&lt; 0.05、**/?&lt; 0.01、 ***;?&lt;〇.01)5。 圖18A-18B係為AcP-IgAN小鼠腎臟中之細胞凋亡試驗結 果:(圖18A)以末端去氧核苷酸轉移酶-相關dUTP缺口末端 標記法(TUNEL)測量腎臟中之細胞凋亡結果,其中最初放 大倍率為400X ;(圖18B)腎臟中有細胞凋亡現象之細胞 數;此些數據係以每組各6隻小鼠所得到之實驗平均值 土SEM表示,其中 */?&lt; 0.05、**/?&lt; 0.01、***/?&lt;0.005。 【主要元件符號說明】 無0 9795 201247612 The mean value of the experiment obtained by each of the 6 mice is ± SEm, where *p &lt; 0.05, **p &lt; 0.01, ***p &lt; 〇.〇〇5. Figures 14A-14F are the results of mRNA and protein expression of Nrf2 in AcP-IgAN mice: (Fig. 14A) Results of mRNA measurement of renal Nrf2 by real-time PCR; (圏14B) Measurement of mRNA of kidney NQ01 by real-time PCR The result of the amount; (Fig. 14C) the result of measuring the amount of mRNA of kidney HO-1 by real-time PCR; (Fig. 14D) the result of measuring the amount of nuclear ΝγΠ in the kidney by ELISA; (Fig. 14Ε) measuring the cell ΗΟ-1 in the kidney by ELISA Results of performance; (14F) GPx activity results in kidneys; these data are expressed as SEM of experimental mean values obtained from 6 mice per group, where *p&lt; 0.05, **/?&lt; 0.01, *** Corpse &lt;〇.〇〇5. Figures 15A-15D are measurements of serum inflammatory factor levels in AcP-IgAN mice; (Figure 15A) IL-6; (Figure 15B) MCP-1; (Figure 15C) IL-Ιβ; (Figure 15D) IL- 18; such data are expressed as SEM of the experimental mean values obtained for each group of 6 mice, wherein * corpse &lt; 0.05, **p &lt; 0.01, ***; ? &lt; 〇.〇〇5.囷16A-16F is a measure of renal NLRP3 inflammatory body activation in AcP-IgAN mice: (Fig. 16A) Measurement of renal NLRP mRNA expression by real-time PCR; (Fig. 16B) Measurement of renal apoptosis factor-1 by real-time PCR mRNA expression level; (Fig. 16C) mRNA expression of renal IL-Ιβ was measured by real-time PCR; (Fig. 16D) mRNA expression of renal IL-18 was measured by real-time PCR; (Fig. 16E) NLRP3 western ink in kidney tissue Point test results; (Fig. 16F) Western blot test results of apoptotic factor-1 (Casp 1) in kidney tissue; among them, Caspl p20 subunit appeared to represent activation, β-actin as internal control histone, 0 · 05, 0·01, ***ρ&lt;〇·〇〇5. 96 201247612 Figures 17A-17F are examples of inhibition of IL-6 and MCP-1 expression in the kidneys of AcP-IgAN mice by a cyclohexenone compound 1 and an increase in NF-κΒ activation: (Fig. 17A) Measurement of NF by tissue immunostaining The result of -κΒ p65, where the initial magnification was 400X, and the numerical series is below the figure; (Fig. 17B) the result of measuring NF-κΒ in the kidney using the ELISA-based TransAM NF-κΒ set; (Fig. 17C) measuring the kidney by real-time PCR The result of the amount of mRNA of MCP-1; (Fig. 17D) the result of measuring the mRNA amount of renal IL-6 by real-time PCR; (Fig. 17E) the result of measuring the protein amount of renal MCP-1 by tissue immunostaining; (Fig. 17F) The results of tissue immunostaining were used to measure the protein content of renal IL-6; the initial magnification was 400X, and the numerical series are shown below. These data are expressed by the mean SEM of the experimental results obtained from each group of 6 mice. Where */7&lt; 0.05, **/?&lt; 0.01, ***;?&lt;〇.01)5. Figures 18A-18B are the results of apoptosis assays in the kidneys of AcP-IgAN mice: (Fig. 18A) Measurement of apoptosis in the kidney by terminal deoxynucleotidyl transferase-related dUTP nick end labeling (TUNEL) As a result, the initial magnification was 400X; (Fig. 18B) the number of cells having apoptosis in the kidney; these data were expressed by the mean SEM of the experimental results obtained from 6 mice in each group, where */? &lt; 0.05, **/?&lt; 0.01, ***/?&lt;0.005. [Main component symbol description] None 0 97

Claims (1)

201247612 七、申請專利範圍· 1. 一種治療一主體之腎絲球硬化症或腎絲球腎炎之 方法,包括: 投予一有效劑量之一環己烯酮化合物'其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,其中,該環己烯酮化合物係具有下列結構:201247612 VII. Scope of Application for Patent 1. A method for treating renal sclerosing or renal glomerulonephritis in a subject, comprising: administering an effective dose of one of the cyclohexenone compounds, a pharmaceutically acceptable salt thereof, a metabolite, a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure: 其中,每一X及Y係各自獨立為氧、nr5、或硫; R係為氫、或CpOKVCs烷基; 每一 R,、R2、及R3係各自獨立為氫、甲基、咬 (CH2)m-CH3 ; R4係為 NR5R6、OR5、〇C(=0)R7、C(=〇)〇r5、c(=〇)r5、 卤素、5或6員環内酯、CVC8烷基、C2-C8烯基、c2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、or5、oc(=o)r7、C(=0)0R5、c(=〇)R5、 Ci-C8炫基、C2-C8稀基、C2-C8块基、C3-C8環烧基、C|-C8 鹵院基、及CrC8烧氧基之取代基所取代; 每一 Rs、及Re係各自獨立為氫、或C丨-C8烷基; R_7係C|-Cg炫基、OR5、或nr5r6 ; m為1-12 ;以及 η為 1-12 » 98 201247612 2. —種治療 主體之腎功能損害或腎絲球損傷 法,包括: 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,其中,該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, nr5, or sulfur; R is hydrogen or CpOKVCs alkyl; each R, R2, and R3 are independently hydrogen, methyl, and bite (CH2) m-CH3; R4 is NR5R6, OR5, 〇C(=0)R7, C(=〇)〇r5, c(=〇)r5, halogen, 5 or 6 membered ring lactone, CVC8 alkyl, C2- a C8 alkenyl group, a c2-C8 alkynyl group, an aryl group, or a glucosyl group, wherein 5 or 6 membered ring lactones, alkyl groups, C2-C8 alkenyl groups, CrC8 alkynyl groups, aryl groups, and glucosyl group-selective One or more selected from the group consisting of NR5R6, or5, oc(=o)r7, C(=0)0R5, c(=〇)R5, Ci-C8 炫, C2-C8 dilute, C2-C8 block, C3 Substituting -C8 cycloalkyl, C|-C8 halogen-based, and CrC8 alkoxy substituent; each Rs, and Re are each independently hydrogen or C丨-C8 alkyl; R_7 is C|- Cg Hyun, OR5, or nr5r6; m is 1-12; and η is 1-12 » 98 201247612 2. A method of treating renal impairment or necrosis of the kidney, including: administering one of the effective doses of the ring a hexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a prodrug thereof thereof A main body, wherein the cyclohexenone compound had the following structure: 其中,每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或烷基; 每一心、R2、及R3係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 NR5R6、or5、oc(=o)r7、c(=o)or5、c(=o)r5、 鹵素、5或6員環内酯、cvc8烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、CrCs烷基、C2-C8 烯基、C2-C8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、0C(=0)R7、c(=o)or5、c(=o)r5、 C|-Cg烧基、C2-C8稀基、C2-C8快基、C3-C8環烧基、Ci_Cg 鹵烷基、及C丨-c8烷氧基之取代基所取代; 每一R5、及係各自獨立為氫、或C|-C8烧基; RACi-Cs烷基、〇r5、或NR5R6 ; m為1-12 ;以及 η為 1.-12。 99 201247612 3. —種提升一主體之腎細胞核因數-E2相關因數2 (Nrf2)活性之方法,包括: 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 趙’其中’該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, NR5, or sulfur; R is hydrogen or an alkyl group; each core, R2, and R3 are each independently hydrogen, methyl, or (CH2)m-CH3 R4 is NR5R6, or5, oc(=o)r7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, cvc8 alkyl, C2-C8 alkenyl, C2 a -C8 alkynyl, aryl, or glucosyl group, wherein 5 or 6 membered ring lactones, CrCs alkyl groups, C2-C8 alkenyl groups, C2-C8 alkynyl groups, aryl groups, and glucosyl group-selective ones Or above selected from NR5R6, OR5, 0C(=0)R7, c(=o)or5, c(=o)r5, C|-Cg alkyl, C2-C8 dilute, C2-C8 fast radical, C3- Substituted by a C8 cycloalkyl, Ci_Cg haloalkyl, and C丨-c8 alkoxy substituent; each R5, and each independently hydrogen, or C|-C8 alkyl; RACi-Cs alkyl, hydrazine R5, or NR5R6; m is 1-12; and η is 1.-12. 99 201247612 3. A method for increasing renal cell nuclear factor-E2 correlation factor 2 (Nrf2) activity of a subject, comprising: administering an effective dose of one cyclohexenone compound, a pharmaceutically acceptable salt thereof, and metabolism thereof a substance, a solvate thereof, or a prodrug thereof to the main Zhao', wherein the cyclohexenone compound has the following structure: 其中’每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CpCOCVCs烷基; 每一 Ri'R2、及R3係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 NR5R6、OR5、〇C(=0)R7、C(=〇)〇R5、C(=〇)R5、 卤素、5或6員環内酯、CVC8烷基、C2-C8烯基、C2-C8块基、 芳基、或葡萄糖基’其中’5或6員環内酯、CrC8烷基、C2_C8 烯基、Cz-Cs炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、0C(=0)R7、C(=0)〇R5、c(=〇)r5、 C!-C8烧基、C2-C8烯基、c2-c8块基、C3-C8環烧基、c 鹵烷基、及C!-C8烷氧基之取代基所取代; 每一 Rs、及R·6係各自獨立為氫、.或c丨-c8烷基; R7係 C|-C8烧基、OR5、或 NR5R_6 ; m為1-12 ;以及 η 為 1-12 〇 100 201247612 4. -種抑制一主體之腎NF-κΒ活性及/或轉化生長因 子(TGF)-〖U蛋白表現之方法,包括: 投予一有效劑量之一環己稀酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體’其中,該環己烯酮化合物係具有下列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or CpCOCVCs alkyl; each Ri'R2, and R3 are each independently hydrogen, methyl, or (CH2)m -CH3 ; R4 is NR5R6, OR5, 〇C(=0)R7, C(=〇)〇R5, C(=〇)R5, halogen, 5 or 6 membered ring lactone, CVC8 alkyl, C2-C8 Alkenyl, C2-C8 block, aryl, or glucosyl' wherein '5 or 6 membered ring lactone, CrC8 alkyl, C2_C8 alkenyl, Cz-Cs alkynyl, aryl, and glucosyl selective One or more selected from the group consisting of NR5R6, OR5, 0C(=0)R7, C(=0)〇R5, c(=〇)r5, C!-C8 alkyl, C2-C8 alkenyl, c2-c8 Substituting a C3-C8 cycloalkyl group, a c-haloalkyl group, and a C!-C8 alkoxy group; each of Rs and R.6 is independently hydrogen, or c丨-c8 alkyl; R7 is C|-C8 alkyl, OR5, or NR5R_6; m is 1-12; and η is 1-12 〇100 201247612 4. Inhibition of a subject's renal NF-κΒ activity and/or transforming growth factor (TGF) - U protein expression method, comprising: administering an effective dose of one of cyclohexanone compounds, pharmaceutically acceptable salts thereof, and metabolism thereof And a solvate thereof, or a prodrug thereof to the host, wherein the cyclohexenone compound has the following structure: 其中’每一 X及Y係各自獨立為氧、nr5、或硫; R係為氫、或(::(=0)(:,-0:8烷基; 每一 R丨、R2、及&amp;係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為NR5R6、OR5、〇C(=0)R7、C(=0)0R5、C(=〇)R5、 鹵素、5或6員環内醋、CVCs烧基、CrC8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Cl_c8烷基、C2_C8 烯基、CrCg炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、or5、0C(=0)R7、c(=〇)〇R5、c(=〇)R5 基、c2-c8稀基、c2-C8块基、&lt;:3-&lt;:8環燒基、C,-C{ 鹵淀基、及Ci-C8烧氧基之取代基所取代; 每一R5、及係各自獨立為氫、或C丨_C8烷基; R7係 C「C8烷基、〇R5、或 nr5r6 ; m為M2 ;以及 η為 1-12 。 101 201247612 之方法,包 一種抑制一主體之ROS/ΝΟ及/或p4 7phox 投予一有效劑量之一環己稀酮化合物、其醫藥上可接 爻之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體’其中’該環己烯酮化合物係具有下列結構:Wherein 'each X and Y are each independently oxygen, nr5, or sulfur; R is hydrogen, or (::(=0)(:,-0:8 alkyl; each R丨, R2, and & Each is independently hydrogen, methyl, or (CH2)m-CH3; R4 is NR5R6, OR5, 〇C(=0)R7, C(=0)0R5, C(=〇)R5, halogen, 5 Or 6-membered ring vinegar, CVCs alkyl, CrC8 alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered cyclic lactone, Cl_c8 alkyl, C2_C8 alkenyl, CrCg alkynyl, The aryl group and the glucose group are selectively selected from the group consisting of NR5R6, or5, 0C(=0)R7, c(=〇)〇R5, c(=〇)R5 group, c2-c8 base group, c2 Substituting -C8 block, &lt;:3-&lt;:8 cycloalkyl, C,-C{halide, and Ci-C8 alkoxy substituent; each R5, and each independently hydrogen Or C丨_C8 alkyl; R7 is C "C8 alkyl, 〇R5, or nr5r6; m is M2; and η is 1-12. 101 201247612, a method of inhibiting a ROS/ΝΟ and// Or p4 7phox is administered as an effective dose of one of the cyclohexanone compounds, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a precursor thereof The drug to the host 'where' the cyclohexenone compound has the following structure: 其中’每一 X及γ係各自獨立為氧、Nr5、或戚; R係為氫、或¢:(=0)0,-(:8炫基; 每一 Ri、R2、及R_3係各自獨立為氫、曱基、或 (CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)r7、c(=〇&gt;〇R5、C(=0)R5、 鹵素、5或6員環内酯、c〗-c8烷基、c2-C8稀基、C2-C8炔基、 芳基、或葡萄糖基’其中,5或6員環内酯、crC8烷基、C2-C8 稀基、C2_C8炔基、芳基、及葡萄糖基係選择性的經一種或 以上選自由 NR5R6、OR5、〇c(=〇)r7、c(=〇)〇R5、C(=0)R5、 烧基、C2-C8稀基、(:2-(:8炔基、c3-C8環炫基、 鹵烧基、及Ci-C8炫氧基之取代基所取代; 每一 R5、及R6係各自獨立為氫、或C1_C8;J^基; R7係 Ci-C8烧基、OR5 ' 或nr5r6 ; m為1 -12 ;以及 η為 1-12 。 102 201247612 6. 一種減少一主體之CD3+/CD69+ T細胞之方法,包 括: 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 截’其中,該環己烯酮化合物係具有下列結構:Wherein 'each X and γ are each independently oxygen, Nr5, or 戚; R is hydrogen, or ¢: (=0)0, -(:8 炫; each Ri, R2, and R_3 are independent Is hydrogen, fluorenyl, or (CH2)m-CH3; R4 is NR5R6, 〇R5, 〇c(=〇)r7, c(=〇&gt;〇R5, C(=0)R5, halogen, 5 or 6-membered ring lactone, c--c8 alkyl, c2-C8 dilute, C2-C8 alkynyl, aryl, or glucosyl' wherein 5 or 6 membered cyclic lactone, crC8 alkyl, C2-C8 One or more selected from the group consisting of NR5R6, OR5, 〇c(=〇)r7, c(=〇)〇R5, C(=0)R5, Substituted by a substituent such as a pyridyl group, a C2-C8 dilute group, a (2-(8-alkynyl group, a c3-C8 cyclohexyl group, a halogen group, and a Ci-C8 methoxy group); each R5, and R6 system Each is independently hydrogen, or C1_C8; J^; R7 is Ci-C8 alkyl, OR5' or nr5r6; m is 1 -12; and η is 1-12. 102 201247612 6. A CD3+/CD69+ reducing a subject A method of T cells, comprising: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, Or a pre-drug thereof to the main section, wherein the cyclohexenone compound has the following structure: 其中’每一X及Υ係各自獨立為氧、nr5、或硫; R係為氫、或C(=0)CrC8烷基; 每一 Ri、R·2、及R_3係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 NR5R6、OR5、〇c(=o)r7、C(=〇)〇R5、C(=0)R5、 _素' 5或6員環内酯、C「C8烧基、c2-c8烯基、c2-c8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、Ci-Cg烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、〇C(=〇)R7、c(=〇)〇R5、C(=0)R5、 C|-C8 燒基、C2-C8:wpr基、c2_c8 块基、C3-C8 環:¾ 基、CVC8 鹵燒基、及Ci-C8烧氧基之取代基所取代; 每一 R5、及R6係各自獨立為氫、或Cl_C8烷基; R?係 Ci-Cs烧基、OR5、或NR5R6 ; m為1-12 ;以及 η為 1-12 。 103 201247612 7. —種提升腎臟中榖胱甘肽過氧化酶(GPx)活性之方 法’包括: 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,其中,該環己烯酮化合物係具有下列結構:Wherein 'each X and oxime are each independently oxygen, nr5, or sulfur; R is hydrogen, or C(=0)CrC8 alkyl; each of Ri, R.2, and R_3 is independently hydrogen, Or, (CH2)m-CH3; R4 is NR5R6, OR5, 〇c(=o)r7, C(=〇)〇R5, C(=0)R5, _素' 5 or 6 membered ring lactone , C"C8 alkyl, c2-c8 alkenyl, c2-c8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered ring lactone, Ci-Cg alkyl, C2-C8 alkenyl, CrC8 alkyne The one, at least one selected from the group consisting of NR5R6, OR5, 〇C(=〇)R7, c(=〇)〇R5, C(=0)R5, C|-C8 is preferably selected from the group consisting of NR5R6, OR5, 〇C(=〇)R7, C(=〇)〇R5, Substituent, C2-C8: wpr group, c2_c8 block group, C3-C8 ring: 3⁄4 group, CVC8 halogen group, and Ci-C8 alkoxy substituent; each R5, and R6 are independently hydrogen Or Cl_C8 alkyl; R? is Ci-Cs alkyl, OR5, or NR5R6; m is 1-12; and η is 1-12. 103 201247612 7. - Raising glutathione peroxidase in the kidney ( The method of GPx) activity comprises: administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, and a solvent thereof Thereof, or a pharmaceutical predecessor to the main body, wherein the cyclohexenone compound had the following structure: 其中’每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CbCOCi-Cs烷基; 每一 R,、R2、及Rs係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 nr5r6、〇r5、〇c(=o)r7、C(=0)0R5、c(=〇)R5、 齒素、5或6員環内醋、Q-Cs院基、C2-C8稀基、C2_C8诀基、 芳基、或葡萄糖基,其中,5或6員環内醋、Ci_C8烷基、C2_C8 烯基、fs炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇r5、oc(=〇)R7、c(=〇)〇R5 c(=〇)R5 CVC8燒基、c2_c8烯基、c2_c8炔基、C3_C8環燒基、C「5C8 齒烧基 '及Ci-C8烷氧基之取代基所取代; 每一R5、及R6係各自獨立為氫、或C丨·C8烷基; R7係 CVC8烷基、〇R5、或NR5R6 ; ni為1 -12 ;以及 η為 1·12 。 8· —種減少一主體之促炎細胞因子 丁&lt;方法,包括: 104 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,其中,該環己烯酮化合物係具有下列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or CbCOCi-Cs alkyl; each R, R2, and Rs are each independently hydrogen, methyl, or ( CH2)m-CH3; R4 is nr5r6, 〇r5, 〇c(=o)r7, C(=0)0R5, c(=〇)R5, dentate, 5 or 6 member vinegar, Q-Cs Affiliation, C2-C8 dilute, C2_C8 decyl, aryl, or glucosyl, wherein 5 or 6 membered vinegar, Ci_C8 alkyl, C2_C8 alkenyl, fs alkynyl, aryl, and glucosyl One or more selected from the group consisting of NR5R6, 〇r5, oc(=〇)R7, c(=〇)〇R5 c(=〇)R5 CVC8 alkyl, c2_c8 alkenyl, c2_c8 alkynyl, C3_C8 cycloalkyl, C "5C8 dentate" and a substituent of Ci-C8 alkoxy group; each R5, and R6 are each independently hydrogen or C丨·C8 alkyl; R7 is CVC8 alkyl, 〇R5, or NR5R6; ni is 1 -12; and η is 1·12. 8. A method for reducing a pro-inflammatory cytokine factor in a subject, comprising: 104 201247612 administering an effective dose of one cyclohexenone compound, its medicine Acceptable salts, metabolites, solvates thereof, or precursors thereof The drug is to the host, wherein the cyclohexenone compound has the following structure: 其中,每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或(:(=0)(ν&lt;:8烷基; 每一 及R3係各自獨立為氩、曱基、或 (CH2)m-CH3 ; R4係為 NR5R6、or5、oc(=o)r7、c(=〇)〇r5、c(=o)r5、 函素、5或6員環内酯、C,-C8炫基、c2-c8烯基、C2-C8快基、 芳基、或葡萄糖基,其中’5或6員環内酯、Cl-C8烷基、C2-C8 烯基、Ca-C8炔基’芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、〇C(=0)R7、c(=〇)〇r5、c(=0)R5、 C1-C8燒基、c2-C8稀基 ' C2-C8块基、c3-c8環烧基、C|-C8 南烧基、及CrC:8烷氧基之取代基所取代; 每一 R5、及R_6係各自獨立為氫、或烧基; 尺7係 Ci-Cg烧基、OR5、或 NR5R6 ; m為1-12 ;以及 η為:卜12。 皰因子包括MCP-1、IL-6、 9.如申請專利範園第8項所述之方法,其中該促炎細 JL-Ιβ、IL-18、或其組合。 105 201247612 10· —種減少腎臟中硫胱胺酸蛋白酶-1表現及/或抑制 腎NLRP3活性之方法,包括: 投予一有效劑量之一環己稀酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,其中,該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, NR5, or sulfur; R is hydrogen, or (:(=0)(ν&lt;:8 alkyl; each and R3 are independently argon, sulfhydryl Or (CH2)m-CH3; R4 is NR5R6, or5, oc(=o)r7, c(=〇)〇r5, c(=o)r5, tropin, 5 or 6-membered ring lactone, C , -C8 leucoyl, c2-c8 alkenyl, C2-C8 fast radical, aryl, or glucosyl, wherein '5 or 6 membered cyclic lactone, Cl-C8 alkyl, C2-C8 alkenyl, Ca-C8 Alkynyl 'aryl, and glucosyl-selective one or more selected from the group consisting of NR5R6, OR5, 〇C(=0)R7, c(=〇)〇r5, c(=0)R5, C1-C8 Substituted by a substituent of a c2-C8 dilute 'C2-C8 block group, a c3-c8 cycloalkyl group, a C|-C8 south alkyl group, and a CrC:8 alkoxy group; each R5, and R_6 system are each substituted Independently hydrogen or calcined; Rule 7 is Ci-Cg alkyl, OR5, or NR5R6; m is 1-12; and η is: Bu 12. Blisters include MCP-1, IL-6, 9. Apply The method of claim 8, wherein the proinflammatory fine JL-Ιβ, IL-18, or a combination thereof. 105 201247612 10· a reduction in the expression and/or inhibition of thiocysteine-1 in the kidney A method of renal NLRP3 activity, comprising: administering an effective dose of one of a cyclohexanone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a prodrug thereof to the subject, wherein the ring The hexenone compound has the following structure: 其中,每一X及Y係各自獨立為氧、nr5、或硫; R係為氫、或¢:(=0)(:,-(:8烷基; 每一 R丨、Rz、及&amp;係各自獨立為氫、曱基、或 (CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇C(=〇)R7、c(=o)or5、c(=o)r5、 卤素、5或6員環内酯、C,-C8烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基,其中’5或6員環内酯、CrCe烷基、C2-C8 稀基、C^-C:8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇C(=〇)R7、C(=0)0R5、C(=0)R5、 CrC8院基、C2-C8稀基、C2-C8炔基、0:3-(:8環烧基、(VC8 鹵烷基、及C,-C8烷氧基之取代基所取代; 每一Rs、及係各自獨立為氫、或CpCs烷基; 烷基、〇R5、或NR5R6 ; m為1 -12 ;以及 η為 1-12 。 11 · 一種減少腎臟中腎NF-kB量之方法,包括: 106 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,其中,該環己烯酮化合物係具有下列結構: r, cmWherein each X and Y system is independently oxygen, nr5, or sulfur; R is hydrogen, or hydrazine: (=0) (:, -(:8 alkyl; each R丨, Rz, and &amp; Each is independently hydrogen, fluorenyl, or (CH2)m-CH3; R4 is NR5R6, 〇R5, 〇C(=〇)R7, c(=o)or5, c(=o)r5, halogen, 5 Or 6-membered ring lactone, C,-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, aryl, or glucosyl, wherein '5 or 6 membered ring lactone, CrCe alkyl, C2-C8 One or more selected from the group consisting of NR5R6, 〇R5, 〇C(=〇)R7, C(=0)0R5, C( =0) R5, CrC8, K2-C8 dilute, C2-C8 alkynyl, 0:3-(:8 cycloalkyl, (VC8 haloalkyl, and C,-C8 alkoxy substituent) Substituted; each Rs, and each independently hydrogen, or CpCs alkyl; alkyl, hydrazine R5, or NR5R6; m is 1 -12; and η is 1-12. 11 · One reduces renal NF-kB in the kidney A method of dosage comprising: 106 201247612 administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a prodrug thereof thereto A main body, wherein the cyclohexenone compound had the following structure: r, cm 其中,每一X及Y係各自獨立為氧、nr5、或硫; R係為氩、或(:(=0)〇ν(:8烷基; 每一 R〗、R·2、及R3係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 NR5R6、OR5、oc(=o)r7、C(=0)0R5、c(=〇)r5、 南素、5或6員環内醋、C丨-Ce炫•基、C2-C8稀基、C2-C8炔基、 芳基、或葡萄糖基,其中,5或6員環内酯、CrCs烷基、c2&gt;&gt;(:8 烯基、C2_CS炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、OR5、0C(=0)R7、C(=0)OR5、c(=〇)r5、 c,-c8烷基、c2-c8烯基、C2-C8炔基、〇:3-(:8環烷基、Ci_C8 鹵烷基、及c丨-c8烷氧基之取代基所取代; 每一R5、及R6係各自獨立為氫、或Crc8烷基; RtSCVCs烷基、OR5、或 NR5R6 ; m為:U12 ;以及 η為1-12 。 12. —種抑制腎臟中細胞凋亡之方法,包括: 107 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 韹,其中,該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, nr5, or sulfur; R is argon or (:(=0)〇ν(:8 alkyl; each R, R·2, and R3 Each is independently hydrogen, methyl, or (CH2)m-CH3; R4 is NR5R6, OR5, oc(=o)r7, C(=0)0R5, c(=〇)r5, N, 5 or 6 In the ring, vinegar, C丨-Ce Hyun, C2-C8, C2-C8 alkynyl, aryl, or glucosyl, wherein 5 or 6 membered cyclic lactone, CrCs alkyl, c2&gt; (8) alkenyl, C2_CS alkynyl, aryl, and glucosyl-selective one or more selected from NR5R6, OR5, 0C(=0)R7, C(=0)OR5, c(=〇)r5 Substituted with a substituent of c, -c8 alkyl, c2-c8 alkenyl, C2-C8 alkynyl, fluorene: 3-(:8-cycloalkyl, Ci_C8 haloalkyl, and c丨-c8 alkoxy; Each R5, and R6 is independently hydrogen or Crc8 alkyl; RtSCVCs alkyl, OR5, or NR5R6; m is: U12; and η is 1-12. 12. Method for inhibiting apoptosis in the kidney Including: 107 201247612 administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or Huang medicament to the main trend, wherein the cyclohexenone compound had the following structure: 其中’每一 X及Y係各自獨立為氧、nr5、或硫; R係為氫、或CpCOCi-Ce烷基; 每一 Ri、R2、及R3係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)r7、c(=〇)OR5、c(=o)r5、 齒素、5或6員環内酯、C「C8烷基、C2-C8烯基、C2-C8炔基、 芳基、或葡萄糖基’其中,5或6員環内酯、CrCs烷基、C2-C8 烯基、CrC8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇c(=0)r7、c(=〇)〇R5、C(=0)R5、 CrC*燒基' C2-C8稀基、C:rC8炔基、c3-c8環炫基、q-Cs 鹵烷基、及Ci-Ce烷氧基之取代基所取代; 每—R5、及Re係各自獨立為氫、或C丨_C8烷基; R?係Cl_C8院基、〇R5、或nr5r6 ; m為1 -12 ;以及 η 為 1 -12 β 或腎絲球腎炎之方法 13 ·—種保護或預防一主趙腎臟罹患腎絲球硬化症及/ 包括: 108 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體’以減少腎臟中TGF-βΙ蛋白之表現程度及膠原蛋白I、ΠΙ 及IV蛋白聚集’其中,該環己烯酮化合物係具有下列結構:Wherein 'each X and Y are each independently oxygen, nr5, or sulfur; R is hydrogen, or CpCOCi-Ce alkyl; each Ri, R2, and R3 are each independently hydrogen, methyl, or (CH2) m-CH3; R4 is NR5R6, 〇R5, 〇c(=〇)r7, c(=〇)OR5, c(=o)r5, dentate, 5 or 6 membered ring lactone, C"C8 alkane a C2-C8 alkenyl group, a C2-C8 alkynyl group, an aryl group, or a glucosyl group, wherein 5 or 6 membered ring lactones, CrCs alkyl groups, C2-C8 alkenyl groups, CrC8 alkynyl groups, aryl groups, and glucose One or more selected from the group consisting of NR5R6, 〇R5, 〇c(=0)r7, c(=〇)〇R5, C(=0)R5, CrC*alkyl-C2-C8, Substituting C: rC8 alkynyl, c3-c8 cyclodextrin, q-Cs haloalkyl, and Ci-Ce alkoxy; each R5, and Re are independently hydrogen, or C丨_C8 Alkyl; R? is Cl_C8, 〇R5, or nr5r6; m is 1 -12; and η is 1 -12 β or glomerulonephritis method 13 ·-protection or prevention of a kidney Ball sclerosis and / Including: 108 201247612 Administration of an effective dose of one of the cyclohexenone compounds, their pharmaceutically acceptable salts, their generation Thereof, solvate thereof, or the precursor drugs to the main body 'to reduce kidney TGF-βΙ proteins levels of expression and collagen I, ΠΙ and IV protein aggregation' wherein the cyclohexenone compound had the following structure: 其中’每一 X及Υ係各自獨立為氧、NR5 '或硫; R係為氫、或CpOKVCe烷基; 每一 R丨、R2、及R3係各自獨立為氫、甲基、或 (CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)R7、c(=o)or5、c(=o)r5、 鹵素、5或6員環内酯、CrCs烧基、c2-c8稀基、C2-C8炔基、 芳基、或葡萄糖基,其中’ 5或6員環内酯、C^-Ce烷基、C2-C8 烯基、C2_CS炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇C(=〇)R7、c(=o)or5、c(=o)r5、 基、c2-c8稀基、c2-c8块基、c3-c8環烧基、(VC8 鹵烧基、及C丨-C8烧氧基之取代基所取代; 每一Rs、及化係各自獨立為氫、或Cl_C8烷基; 1^7係€;1-08烧基、〇115、或&gt;11151^6; m為1-12 ;以及 η 為:1-12。 14. 一種治療一主體之局部性腎絲球硬化症(FsgS)2 方法,包括: 109 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,以(i)增加Nrf2活性及/或(ii)抑制腎臟中NF-icB相關發 炎反應及TGF-βΙ誘導之纖維化,其中,該環己烯酮化合物 係具有下列結構:Wherein 'each X and oxime are each independently oxygen, NR5' or sulfur; R is hydrogen or CpOKVCe alkyl; each R丨, R2, and R3 are each independently hydrogen, methyl, or (CH2) m-CH3 ; R4 is NR5R6, 〇R5, 〇c(=〇)R7, c(=o)or5, c(=o)r5, halogen, 5 or 6 membered ring lactone, CrCs alkyl, c2- C8 dilute, C2-C8 alkynyl, aryl, or glucosyl, wherein '5 or 6 membered ring lactone, C^-Ce alkyl, C2-C8 alkenyl, C2_CS alkynyl, aryl, and glucosyl Selectively one or more selected from the group consisting of NR5R6, 〇R5, 〇C(=〇)R7, c(=o)or5, c(=o)r5, yl, c2-c8, and c2-c8 And a substituent of a c3-c8 cycloalkyl group, (a VC8 halogen group, and a C丨-C8 alkoxy group; each Rs and a system are independently hydrogen or a Cl_C8 alkyl group; 1^7 is a € 1-08 alkyl, 〇115, or >11151^6; m is 1-12; and η is: 1-12. 14. A method for treating a subject with local squamous sclerosis (FsgS) 2 Including: 109 201247612 administering an effective dose of one of cyclohexenone compounds, pharmaceutically acceptable salts thereof, metabolites thereof, and solvation thereof Or a pre-drug to the subject, (i) increasing Nrf2 activity and/or (ii) inhibiting NF-icB-related inflammatory response in the kidney and TGF-βΙ-induced fibrosis, wherein the cyclohexenone compound is Has the following structure: 其中,每一X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CbCOCi-Cs烷基; 每 Ri、R2、及R_3係各自獨立為氮、曱基、或 (CH2)m-CH3 ; R4係為 NR5R6、〇R5、〇c(=〇)r7 ' c(=〇)〇r5、c(=〇)R5、 函素、5或6員環内醋、Cl_(:8烧基、c2_C8稀基、c2_C8快基、 芳基、或葡萄糖基,其中,5或6員環内酯、Ci_C8烷基、C2_c8 烯基、Cz-C8炔基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5' 〇c(=〇)r7、c卜〇)〇R5、c(=〇)r5、 c,-c8烷基、c2-c8烯基、c2-C8炔基、c3 c8環烷基、C|_C8 鹵烷基、及Ci-C8烷氧基之取代基所取代; 每一R5、及R0係各自獨立為氫、或烷基; 炫基、〇r5、或 nr5r6 ; m為1 -12 ;以及 η 為 1 -12。 15. -種治療-主體之腎絲球腎炎之方法,包括: 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體,以⑴抑制腎NLRP3發炎體活化及/或(Π)抑制丁細胞活 性增加,其中,該環己烯酮化合物係具有下列結構:Wherein each X and Y system is independently oxygen, NR5, or sulfur; R is hydrogen or CbCOCi-Cs alkyl; each of Ri, R2, and R_3 is independently nitrogen, sulfhydryl, or (CH2) m-CH3 ; R4 is NR5R6, 〇R5, 〇c(=〇)r7 'c(=〇)〇r5, c(=〇)R5, pheromone, 5 or 6 member vinegar, Cl_(:8 An alkyl group, a c2_C8 dilute group, a c2_C8 fast group, an aryl group, or a glucosyl group, wherein 5 or 6 membered ring lactones, Ci_C8 alkyl groups, C2_c8 alkenyl groups, Cz-C8 alkynyl groups, aryl groups, and glucosyl group selection One or more selected from the group consisting of NR5R6, 〇R5' 〇c(=〇)r7, c 〇)〇R5, c(=〇)r5, c,-c8 alkyl, c2-c8 alkenyl, c2- Substituted by a C8 alkynyl group, a c3 c8 cycloalkyl group, a C|_C8 haloalkyl group, and a Ci-C8 alkoxy group; each of R5 and R0 is independently hydrogen or an alkyl group; R5, or nr5r6; m is 1 -12; and η is 1 -12. 15. A method of treating a subject-derived glomerulonephritis comprising: 201247612 administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, a metabolite thereof, a solvate thereof, or a precursor thereof The drug is administered to the host to (1) inhibit renal NLRP3 inflammatory body activation and/or (Π) inhibit the increase in butyl cell activity, wherein the cyclohexenone compound has the following structure: 其中’每一 X及Y係各自獨立為氧、NR5、或硫; R係為氫、或CpC^CVCs烷基; 每一 R〗、R2、及R3係各自獨立為氫、曱基、或 (cn2)m-CR3; R4係為 NR5R6、〇R5、〇C(=〇)R7、C(=0)〇R5、c(=〇)r5、 彘素、5或6員環内酯、CVC8烷基、C2-C8烯基、C2-c8炔基、 芳基、或葡萄糖基’其中,5或6員環内酯、C〗-C8烷基、c2-C8 稀基、Ci-C:8块基、芳基、及葡萄糖基係選擇性的經一種或 以上選自由 NR5R6、〇R5、〇c(=〇)R7、C(=0)0R5、c(=〇)r5、 &lt;^-0:8说基、C2-C8稀基、c2-C8炔基、C3-C8環提基、CVCs 卤烷基、及C「C8烷氧基之取代基所取代; 每一Rs、及厌6係各自獨立為氫、或CrC8烷基; R7係 C丨-C8烧基、〇r5、或 nr5r6 ; m為:1·12 ;以及 η為〗-12。 16. —種減緩一主體之免疫球蛋白a型腎絲球腎炎 (IgAN)之方法,包括: 201247612 投予一有效劑量之一環己烯酮化合物、其醫藥上可接 受之鹽類、其代謝物、其溶劑化物、或其前趨藥物至該主 體’其中’該環己烯酮化合物係具有下列結構:Wherein 'each X and Y are each independently oxygen, NR5, or sulfur; R is hydrogen, or CpC^CVCs alkyl; each R, R2, and R3 are each independently hydrogen, sulfhydryl, or ( Cn2)m-CR3; R4 is NR5R6, 〇R5, 〇C(=〇)R7, C(=0)〇R5, c(=〇)r5, alizarin, 5 or 6 membered ring lactone, CVC8 alkane , C2-C8 alkenyl, C2-c8 alkynyl, aryl, or glucosyl' wherein 5 or 6 membered cyclic lactone, C-C8 alkyl, c2-C8 dilute, Ci-C: 8 The one, at least one selected from the group consisting of NR5R6, 〇R5, 〇c(=〇)R7, C(=0)0R5, c(=〇)r5, &lt;^-0 :8, substituted by a C2-C8 dilute group, a C2-C8 alkynyl group, a C3-C8 cyclopentyl group, a CVCs haloalkyl group, and a C"C8 alkoxy group; each Rs, and an anaesthetic 6 system Each is independently hydrogen or a CrC8 alkyl group; R7 is a C丨-C8 alkyl group, 〇r5, or nr5r6; m is: 1·12; and η is 〖-12. 16. - an immunoglobulin that slows down a subject A method of type A glomerulonephritis (IgAN), comprising: 201247612 administering an effective dose of one of a cyclohexenone compound, a pharmaceutically acceptable salt thereof, and a metabolite thereof Solvate thereof, or a pharmaceutical predecessor to the main body of 'where' the cyclohexenone compound had the following structure: 其中’每一 X及Y係各自獨立為氧、nr5、或硫; R係為氫、或c(=0)cvc8烷基; 每一 Ri、R2、及R3係各自獨立為氩、曱基、成 (CH2)m-CH3 ; R4係為 NR5R6、OR5、oc(=o)r7、c(=o)or5、c(=0)R5、 鹵素、5或6員環内酯、烧基、c2-c8稀基、C2-C8炔基, 芳基、或葡萄糖基,其中,5或6員環内酯、C,-C8烷基、C2-C8 烯基、Cz-C8炔基、芳基、及葡萄糖基係選擇性的經一種成 以上選自由 NR5R6、or5、〇c(=o)r7、c(=o)or5、C(=0)R5、 Crc8烷基、c2-c8烯基、C2-C8炔基、C3-C8環烷基、CrCs 鹵烷基、及c丨-c8烷氧基之取代基所取代; 每一R5、及R6係各自獨立為氫、或Crc8烷基; R7係Ci-C8烷基、OR5、或NR5R6 ; m為1-12 ;以及 η為 1-12 。 17.如申請專利範圍第1項所述之方法,其中該腎絲球 硬化症係為局部性腎絲球硬化症(FSGS)或結節性腎絲球硬 化症。 112 201247612 18. 如申請專利範圍第1項所述之方法,其中該腎絲球 硬化症係為局部性腎絲球硬化症(FSGS)。 19. 如申請專利範圍第1項所述之方法,其中該腎絲球 腎炎係為免疫球蛋白A型腎絲球腎炎(IgAN) » 20. 如申請專利範圍第1項所述之方法,其中該環己烯 酮化合物係防止氧化壓力。 21. 如申請專利範圍第20項所述之方法,其中係透過減 少TGF-β】及胞外基質蛋白表現以防止氧化壓力。 22. 如申請專利範圍第1項所述之方法,其中該環己烯 闕化合物係減少該主體中之CD3+/CD69+ T細胞及促炎細胞 因子。 23·如申請專利範圍第22項所述之方法,其中該促炎細 胞因子包括MCP-1、IL-6、IL-Ιβ、IL-18、或其組合。 24, 如申請專利範圍第2項所述之方法,其中該腎絲球 損傷包括上皮增生損傷(EPHL)。 25. 如申請專利範圍第1至24項任一項所述之方法,其 中該化合物係分離自牛樟芝。 26’如申凊專利範圍第1至25項任一項所述之方法,其 中R係為氫、或c(=o)ch3。 27·如申請專利範圍第1至26項任一項所述之方法,其 中Ri係為氫、或甲基。 、 28.如申請專利範圍第1至27項任一項所述之方法,豆 中R2係為氣、或甲基。 八 113 201247612 29. 如申请專利範圍第1至28項任一項所述之方法 中 R4係為 CH2CH=C(CH3)2。 30. 如申請專利範圍第1至29項任一項所述之方法Wherein 'each X and Y are each independently oxygen, nr5, or sulfur; R is hydrogen, or c(=0)cvc8 alkyl; each of Ri, R2, and R3 is independently argon, sulfhydryl, (CH2)m-CH3; R4 is NR5R6, OR5, oc(=o)r7, c(=o)or5, c(=0)R5, halogen, 5 or 6 membered ring lactone, alkyl group, c2 a -c8 dilute group, a C2-C8 alkynyl group, an aryl group, or a glucosyl group, wherein 5 or 6 membered ring lactones, C,-C8 alkyl groups, C2-C8 alkenyl groups, Cz-C8 alkynyl groups, aryl groups, And a glucose-based selective one or more selected from the group consisting of NR5R6, or5, 〇c(=o)r7, c(=o)or5, C(=0)R5, Crc8 alkyl, c2-c8 alkenyl, C2 Substituted by a substituent of -C8 alkynyl, C3-C8 cycloalkyl, CrCs haloalkyl, and c丨-c8 alkoxy; each R5, and R6 are each independently hydrogen or Crc8 alkyl; R7 Ci-C8 alkyl, OR5, or NR5R6; m is 1-12; and η is 1-12. 17. The method of claim 1, wherein the glomerulosclerosis is local glomerulosclerosis (FSGS) or nodular glomerular sclerosis. The method of claim 1, wherein the glomerulosclerosis is localized glomerulosclerosis (FSGS). 19. The method according to claim 1, wherein the glomerulonephritis is an immunoglobulin type A glomerulonephritis (IgAN). The method of claim 1, wherein The cyclohexenone compound prevents oxidation stress. 21. The method of claim 20, wherein the oxidative stress is prevented by reducing TGF-[beta] and extracellular matrix protein expression. 22. The method of claim 1, wherein the cyclohexene oxime compound reduces CD3+/CD69+ T cells and pro-inflammatory cytokines in the subject. The method of claim 22, wherein the pro-inflammatory cytokine comprises MCP-1, IL-6, IL-Ιβ, IL-18, or a combination thereof. [24] The method of claim 2, wherein the renal sac damage comprises epithelial hyperplasia (EPHL). 25. The method of any one of claims 1 to 24, wherein the compound is isolated from Antrodia camphorata. The method of any one of claims 1 to 25, wherein R is hydrogen or c(=o)ch3. The method of any one of claims 1 to 26, wherein the Ri is hydrogen or a methyl group. 28. The method according to any one of claims 1 to 27, wherein R2 in the bean is gas or methyl. </ RTI> </ RTI> </ RTI> </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; </ RTI> <RTIgt; 30. The method of any one of claims 1 to 29 31.如申請專利範圍第1至30項任一項所述之方法 中該主體係為人類。 八、圖式(請見下頁):The method according to any one of claims 1 to 30, wherein the main system is a human. Eight, schema (see next page):
TW101100587A 2011-01-21 2012-01-06 Compositions for treating kidney disorders TWI452032B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161435201P 2011-01-21 2011-01-21
US201161544910P 2011-10-07 2011-10-07

Publications (2)

Publication Number Publication Date
TW201247612A true TW201247612A (en) 2012-12-01
TWI452032B TWI452032B (en) 2014-09-11

Family

ID=46516419

Family Applications (1)

Application Number Title Priority Date Filing Date
TW101100587A TWI452032B (en) 2011-01-21 2012-01-06 Compositions for treating kidney disorders

Country Status (12)

Country Link
US (2) US20150306047A1 (en)
EP (1) EP2667864A4 (en)
JP (1) JP6068361B2 (en)
KR (1) KR20140097970A (en)
CN (1) CN103458884A (en)
AP (1) AP2013007056A0 (en)
BR (1) BR112013018544A2 (en)
CA (1) CA2824352A1 (en)
EA (1) EA201370163A1 (en)
MX (1) MX2013008410A (en)
TW (1) TWI452032B (en)
WO (1) WO2012100228A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI469961B (en) * 2011-12-30 2015-01-21 Golden Biotechnology Corp Usage of compound for preparing drugs for treating diabetes

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9249117B2 (en) * 2014-05-20 2016-02-02 Hui Ling Tseng Use of compounds from Antrodia camphorata in manufacturing medicaments for treating kidney diseases
CN105311005A (en) * 2014-06-10 2016-02-10 曾卉菱 Application of antrodia camphorata compound in preparation of drug for treating kidney diseases
CN105287448A (en) * 2014-06-10 2016-02-03 曾卉菱 Application of antrodia camphorata compound in preparation of drugs for treating kidney diseases
CN105267204A (en) * 2014-06-10 2016-01-27 曾卉菱 Application of antrodia camphorata compound to prepare medicines treating kidney diseases
CN108374029B (en) * 2018-03-12 2020-11-06 江南大学 Method for promoting antrodia camphorata liquid fermentation to produce Antrodin C

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE513823T1 (en) * 2004-08-17 2011-07-15 Simpson Biotech Co Ltd MIXTURE AND COMPOUNDS OF MYCELIA OF ANTRODIA CAMPHORATA AND THEIR USE
TW200829234A (en) * 2007-01-08 2008-07-16 Golden Biotechnology Corp Antrodia camphorata isophorone extract
TW201034657A (en) * 2007-01-08 2010-10-01 Golden Biotechnology Corp Cyclohexene compound
TWI394572B (en) * 2007-06-12 2013-05-01 Golden Biotechnology Corp Application of Cynanchum aurantii Cyclohexenone Compounds in the Preparation of Drugs for the Treatment of Autoimmune Diseases
TWI394573B (en) * 2007-06-14 2013-05-01 Golden Biotechnology Corp Application of Cynanchum auranthone Cyclohexenone Compounds in the Preparation of Drugs for Liver Protection
TWI394574B (en) * 2007-06-14 2013-05-01 Golden Biotechnology Corp Application of Cynanchum auranthone Cyclohexenone Compounds in the Preparation of Drugs for Mitigating Physiological Fatigue
JP5203644B2 (en) * 2007-07-13 2013-06-05 國鼎生物科技股▲ふん▼有限公司 Compounds derived from Benicus mushroom for use in the treatment of autoimmune diseases
CN101357883B (en) * 2007-07-30 2012-09-26 国鼎生物科技股份有限公司 Antrodia camphoratea pimelie kelone compound for treating autoimmune disease and medicine composition
TW200918498A (en) * 2007-10-19 2009-05-01 Golden Biotechnology Corp Novel compound isolated from Antrodia camphorate extracts
CN101417934B (en) * 2007-10-24 2012-04-18 国鼎生物科技股份有限公司 New compounds separated from Antrodia camphorate extract

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI469961B (en) * 2011-12-30 2015-01-21 Golden Biotechnology Corp Usage of compound for preparing drugs for treating diabetes

Also Published As

Publication number Publication date
EP2667864A2 (en) 2013-12-04
WO2012100228A3 (en) 2012-11-22
JP2014511354A (en) 2014-05-15
US20150306047A1 (en) 2015-10-29
KR20140097970A (en) 2014-08-07
TWI452032B (en) 2014-09-11
BR112013018544A2 (en) 2016-10-18
AP2013007056A0 (en) 2013-08-31
US10406120B2 (en) 2019-09-10
CA2824352A1 (en) 2012-07-26
WO2012100228A2 (en) 2012-07-26
CN103458884A (en) 2013-12-18
MX2013008410A (en) 2014-09-08
US20160184239A1 (en) 2016-06-30
JP6068361B2 (en) 2017-01-25
EA201370163A1 (en) 2014-11-28
EP2667864A4 (en) 2014-06-25

Similar Documents

Publication Publication Date Title
JP5247469B2 (en) Inhibition of inflammatory cytokine production using tanshinone
JP7169693B2 (en) Treatment of respiratory diseases
US10406120B2 (en) Methods and compositions for treating kidney disorders
JP2014530242A (en) Topical formulation of chemerin C15 peptide for the treatment of skin diseases
EP2782563B1 (en) Tetrahydrocannabivarin for use in the treatment of intestinal inflammatory diseases
Gao et al. Nicotine-induced autophagy via AMPK/mTOR pathway exerts protective effect in colitis mouse model
EP3666258B1 (en) Method of treating prader-willi syndrome
JP6619361B2 (en) Use of ginsenoside M1 to treat IgA nephropathy
TWI469961B (en) Usage of compound for preparing drugs for treating diabetes
JP2023516284A (en) Compositions and therapeutic uses of cannabidiol
TW201249426A (en) Methods and compositions for treating brain cancer
TWI544921B (en) Use of osthole for manufacturing composition for treating focal segmental glomerulosclerosis
TWI468184B (en) Pharmaceutical composition and medical cosmetics
JP2024519342A (en) Compositions for treating autoimmune, alloimmune, inflammatory, and mitochondrial conditions and uses thereof
WO2014187185A1 (en) Use of pharmaceutical composition in treating dermatitis and eczema
CN103284982A (en) Methods and compositions for treating cancer metastasis
TWI469784B (en) Therapeutic compositoin for treating cancers
CN111212635A (en) Topical compositions
CN111212631A (en) Topical compositions and methods of treatment
TW201417800A (en) Usage of compound for preparing composition for treating, modifying and managing bone cancer pain
CN113768943B (en) Use of TLR4 pathway inhibitors in the manufacture of a medicament
Zhong et al. Similarities and differences between embryonic implantation and CTC invasion: Exploring the roles of abortifacients in cancer metastasis chemoprevention
AU2012207083A1 (en) Methods and compositions for treating kidney disorders
TW202128200A (en) Methods of inducing or enhancing farnesoid x receptor (fxr)-mediated transcriptional response
CN117257790A (en) Application of apigenin in preparation of medicine for treating psoriasis